1
|
Baraniuk JN, Eaton-Fitch N, Marshall-Gradisnik S. Meta-analysis of natural killer cell cytotoxicity in myalgic encephalomyelitis/chronic fatigue syndrome. Front Immunol 2024; 15:1440643. [PMID: 39483457 PMCID: PMC11524851 DOI: 10.3389/fimmu.2024.1440643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 09/09/2024] [Indexed: 11/03/2024] Open
Abstract
Reduced natural killer (NK) cell cytotoxicity is the most consistent immune finding in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). Meta-analysis of the published literature determined the effect size of the decrement in ME/CFS. Databases were screened for papers comparing NK cell cytotoxicity in ME/CFS and healthy controls. A total of 28 papers and 55 effector:target cell ratio (E:T) data points were collected. Cytotoxicity in ME/CFS was significantly reduced to about half of healthy control levels, with an overall Hedges' g of 0.96 (0.75-1.18). Heterogeneity was high but was explained by the range of E:T ratios, different methods, and potential outliers. The outcomes confirm reproducible NK cell dysfunction in ME/CFS and will guide studies using the NK cell model system for pathomechanistic investigations. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42024542140.
Collapse
Affiliation(s)
- James N. Baraniuk
- Department of Medicine, Georgetown University, Washington, DC, United States
- National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia
| | - Natalie Eaton-Fitch
- National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia
| | - Sonya Marshall-Gradisnik
- National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia
| |
Collapse
|
2
|
Cunha I, Latron E, Bauer S, Sage D, Griffié J. Machine learning in microscopy - insights, opportunities and challenges. J Cell Sci 2024; 137:jcs262095. [PMID: 39465533 DOI: 10.1242/jcs.262095] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024] Open
Abstract
Machine learning (ML) is transforming the field of image processing and analysis, from automation of laborious tasks to open-ended exploration of visual patterns. This has striking implications for image-driven life science research, particularly microscopy. In this Review, we focus on the opportunities and challenges associated with applying ML-based pipelines for microscopy datasets from a user point of view. We investigate the significance of different data characteristics - quantity, transferability and content - and how this determines which ML model(s) to use, as well as their output(s). Within the context of cell biological questions and applications, we further discuss ML utility range, namely data curation, exploration, prediction and explanation, and what they entail and translate to in the context of microscopy. Finally, we explore the challenges, common artefacts and risks associated with ML in microscopy. Building on insights from other fields, we propose how these pitfalls might be mitigated for in microscopy.
Collapse
Affiliation(s)
- Inês Cunha
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Tomtebodavägen 23, 171 65 Solna, Sweden
| | - Emma Latron
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Tomtebodavägen 23, 171 65 Solna, Sweden
| | - Sebastian Bauer
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Tomtebodavägen 23, 171 65 Solna, Sweden
| | - Daniel Sage
- Biomedical Imaging Group and EPFL Center for Imaging, École Polytechnique, Rte Cantonale, 1015 Lausanne, Switzerland
| | - Juliette Griffié
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Tomtebodavägen 23, 171 65 Solna, Sweden
| |
Collapse
|
3
|
Hammer Q, Perica K, Mbofung RM, van Ooijen H, Martin KE, Momayyezi P, Varady E, Pan Y, Jelcic M, Groff B, Abujarour R, Krokeide SZ, Lee T, Williams A, Goodridge JP, Valamehr B, Önfelt B, Sadelain M, Malmberg KJ. Genetic ablation of adhesion ligands mitigates rejection of allogeneic cellular immunotherapies. Cell Stem Cell 2024; 31:1376-1386.e8. [PMID: 38981470 DOI: 10.1016/j.stem.2024.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 04/10/2024] [Accepted: 06/14/2024] [Indexed: 07/11/2024]
Abstract
Allogeneic cellular immunotherapies hold promise for broad clinical implementation but face limitations due to potential rejection of donor cells by the host immune system. Silencing of beta-2 microglobulin (B2M) expression is commonly employed to evade T cell-mediated rejection by the host, although the absence of B2M is expected to trigger missing-self responses by host natural killer (NK) cells. Here, we demonstrate that genetic deletion of the adhesion ligands CD54 and CD58 in B2M-deficient chimeric antigen receptor (CAR) T cells and multi-edited induced pluripotent stem cell (iPSC)-derived CAR NK cells reduces their susceptibility to rejection by host NK cells in vitro and in vivo. The absence of adhesion ligands limits rejection in a unidirectional manner in B2M-deficient and B2M-sufficient settings without affecting the antitumor functionality of the engineered donor cells. Thus, these data suggest that genetic ablation of adhesion ligands effectively alleviates rejection by host immune cells, facilitating the implementation of universal immunotherapy.
Collapse
Affiliation(s)
- Quirin Hammer
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden.
| | - Karlo Perica
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA; Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Cell Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Hanna van Ooijen
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Solna, Sweden
| | - Karen E Martin
- Precision Immunotherapy Alliance, Institute for Cancer Research, University of Oslo, Oslo, Norway; Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Pouria Momayyezi
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | | | - Yijia Pan
- Fate Therapeutics, Inc., San Diego, CA, USA
| | | | | | | | - Silje Z Krokeide
- Precision Immunotherapy Alliance, Institute for Cancer Research, University of Oslo, Oslo, Norway; Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Tom Lee
- Fate Therapeutics, Inc., San Diego, CA, USA
| | | | | | | | - Björn Önfelt
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden; Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Solna, Sweden
| | - Michel Sadelain
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Karl-Johan Malmberg
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden; Precision Immunotherapy Alliance, Institute for Cancer Research, University of Oslo, Oslo, Norway; Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
4
|
Dixon KJ, Snyder KM, Khaw M, Hullsiek R, Davis ZB, Matson AW, Shirinbak S, Hancock B, Bjordahl R, Hosking M, Miller JS, Valamehr B, Wu J, Walcheck B. iPSC-derived NK cells expressing high-affinity IgG Fc receptor fusion CD64/16A to mediate flexible, multi-tumor antigen targeting for lymphoma. Front Immunol 2024; 15:1407567. [PMID: 39100677 PMCID: PMC11294090 DOI: 10.3389/fimmu.2024.1407567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 06/21/2024] [Indexed: 08/06/2024] Open
Abstract
Introduction NK cells can mediate tumor cell killing by natural cytotoxicity and by antibody-dependent cell-mediated cytotoxicity (ADCC), an anti-tumor mechanism mediated through the IgG Fc receptor CD16A (FcγRIIIA). CD16A polymorphisms conferring increased affinity for IgG positively correlate with clinical outcomes during monoclonal antibody therapy for lymphoma, linking increased binding affinity with increased therapeutic potential via ADCC. We have previously reported on the FcγR fusion CD64/16A consisting of the extracellular region of CD64 (FcγRI), a high-affinity Fc receptor normally expressed by myeloid cells, and the transmembrane/cytoplasmic regions of CD16A, to create a highly potent and novel activating fusion receptor. Here, we evaluate the therapeutic potential of engineered induced pluripotent stem cell (iPSC)-derived NK (iNK) cells expressing CD64/16A as an "off-the-shelf", antibody-armed cellular therapy product with multi-antigen targeting potential. Methods iNK cells were generated from iPSCs engineered to express CD64/16A and an interleukin (IL)-15/IL-15Rα fusion (IL-15RF) protein for cytokine independence. iNK cells and peripheral blood NK cells were expanded using irradiated K562-mbIL21-41BBL feeder cells to examine in in vitro and in vivo assays using the Raji lymphoma cell line. ADCC was evaluated in real-time by IncuCyte assays and using a xenograft mouse model with high circulating levels of human IgG. Results Our data show that CD64/16A expressing iNK cells can mediate potent anti-tumor activity against human B cell lymphoma. In particular, (i) under suboptimal conditions, including low antibody concentrations and low effector-to-target ratios, iNK-CD64/16A cells mediate ADCC, (ii) iNK-CD64/16A cells can be pre-loaded with tumor-targeting antibodies (arming) to elicit ADCC, (iii) armed iNK-CD64/16A cells can be repurposed with additional antibodies to target new tumor antigens, and (iv) cryopreserved, armed iNK-CD64/16A are capable of sustained ADCC in a tumor xenograft model under saturating levels of human IgG. Discussion iNK-CD64/16A cells allow for a flexible use of antibodies (antibody arming and antibody targeting), and an "off-the-shelf" platform for multi-antigen recognition to overcome limitations of adoptive cell therapies expressing fixed antigen receptors leading to cancer relapse due to antigen escape variants.
Collapse
Affiliation(s)
- Kate J. Dixon
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Kristin M. Snyder
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Melissa Khaw
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Robert Hullsiek
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Zachary B. Davis
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Anders W. Matson
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | | | | | | | | | - Jeffrey S. Miller
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
- Center for Immunology, University of Minnesota, Minneapolis, MN, United States
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | | | - Jianming Wu
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - Bruce Walcheck
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
- Center for Immunology, University of Minnesota, Minneapolis, MN, United States
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
5
|
Yeh M, Salazar-Cavazos E, Krishnan A, Altan-Bonnet G, DeVoe DL. Probing T-cell activation in nanoliter tumor co-cultures using membrane displacement trap arrays. Integr Biol (Camb) 2024; 16:zyae014. [PMID: 39074471 PMCID: PMC11286267 DOI: 10.1093/intbio/zyae014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 06/26/2024] [Accepted: 07/18/2024] [Indexed: 07/31/2024]
Abstract
Immune responses against cancer are inherently stochastic, with small numbers of individual T cells within a larger ensemble of lymphocytes initiating the molecular cascades that lead to tumor cytotoxicity. A potential source of this intra-tumor variability is the differential ability of immune cells to respond to tumor cells. Classical microwell co-cultures of T cells and tumor cells are inadequate for reliably culturing and analyzing low cell numbers needed to probe this variability, and have failed in recapitulating the heterogeneous small domains observed in tumors. Here we leverage a membrane displacement trap array technology that overcomes limitations of conventional microwell plates for immunodynamic studies. The microfluidic platform supports on-demand formation of dense nanowell cultures under continuous perfusion reflecting the tumor microenvironment, with real-time monitoring of T cell proliferation and activation within each nanowell. The system enables selective ejection of cells for profiling by fluorescence activated cell sorting, allowing observed on-chip variability in immune response to be correlated with off-chip quantification of T cell activation. The technology offers new potential for probing the molecular origins of T cell heterogeneity and identifying specific cell phenotypes responsible for initiating and propagating immune cascades within tumors. Insight Box Variability in T cell activation plays a critical role in the immune response against cancer. New tools are needed to unravel the mechanisms that drive successful anti-tumor immune response, and to support the development of novel immunotherapies utilizing rare T cell phenotypes that promote effective immune surveillance. To this end, we present a microfluidic cell culture platform capable of probing differential T cell activation in an array of nanoliter-scale wells coupled with off-chip cell analysis, enabling a high resolution view of variable immune response within tumor / T cell co-cultures containing cell ensembles orders of magnitude smaller than conventional well plate studies.
Collapse
Affiliation(s)
- Michael Yeh
- Department of Mechanical Engineering, University of Maryland, College Park, MD 20742, United States
- Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD 20742, United States
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | | | - Anagha Krishnan
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Grégoire Altan-Bonnet
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Don L DeVoe
- Department of Mechanical Engineering, University of Maryland, College Park, MD 20742, United States
- Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD 20742, United States
| |
Collapse
|
6
|
Hammer Q, Perica K, van Ooijen H, Mbofung R, Momayyezi P, Varady E, Martin KE, Pan Y, Jelcic M, Groff B, Abujarour R, Krokeide S, Lee T, Williams A, Goodridge JP, Valamehr B, Önfelt B, Sadelain M, Malmberg KJ. Genetic ablation of adhesion ligands averts rejection of allogeneic immune cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.09.557143. [PMID: 37873468 PMCID: PMC10592662 DOI: 10.1101/2023.10.09.557143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Allogeneic cell therapies hold promise for broad clinical implementation, but face limitations due to potential rejection by the recipient immune system. Silencing of beta-2-microglobulin ( B2M ) expression is commonly employed to evade T cell-mediated rejection, although absence of B2M triggers missing-self responses by recipient natural killer (NK) cells. Here, we demonstrate that deletion of the adhesion ligands CD54 and CD58 on targets cells robustly dampens NK cell reactivity across all sub-populations. Genetic deletion of CD54 and CD58 in B2M -deficient allogeneic chimeric antigen receptor (CAR) T and multi-edited induced pluripotent stem cell (iPSC)-derived NK cells reduces their susceptibility to rejection by NK cells in vitro and in vivo without affecting their anti-tumor effector potential. Thus, these data suggest that genetic ablation of adhesion ligands effectively alleviates rejection of allogeneic immune cells for immunotherapy.
Collapse
|
7
|
Sandoz PA, Kuhnigk K, Szabo EK, Thunberg S, Erikson E, Sandström N, Verron Q, Brech A, Watzl C, Wagner AK, Alici E, Malmberg KJ, Uhlin M, Önfelt B. Modulation of lytic molecules restrain serial killing in γδ T lymphocytes. Nat Commun 2023; 14:6035. [PMID: 37758698 PMCID: PMC10533871 DOI: 10.1038/s41467-023-41634-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
γδ T cells play a pivotal role in protection against various types of infections and tumours, from early childhood on and throughout life. They consist of several subsets characterised by adaptive and innate-like functions, with Vγ9Vδ2 being the largest subset in human peripheral blood. Although these cells show signs of cytotoxicity, their modus operandi remains poorly understood. Here we explore, using live single-cell imaging, the cytotoxic functions of γδ T cells upon interactions with tumour target cells with high temporal and spatial resolution. While γδ T cell killing is dominated by degranulation, the availability of lytic molecules appears tightly regulated in time and space. In particular, the limited co-occurrence of granzyme B and perforin restrains serial killing of tumour cells by γδ T cells. Thus, our data provide new insights into the cytotoxic arsenal and functions of γδ T cells, which may guide the development of more efficient γδ T cell based adoptive immunotherapies.
Collapse
Affiliation(s)
- Patrick A Sandoz
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden.
| | - Kyra Kuhnigk
- Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Edina K Szabo
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Sarah Thunberg
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Elina Erikson
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Niklas Sandström
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Quentin Verron
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Andreas Brech
- Cancell, Centre for Cancer Cell Reprogramming, Department for Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Biosciences, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University, Oslo, Norway
| | - Carsten Watzl
- Department for Immunology, Leibniz Research Centre for Working Environment and Human Factors, TU Dortmund, Dortmund, Germany
| | - Arnika K Wagner
- Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Evren Alici
- Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Karl-Johan Malmberg
- Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Michael Uhlin
- CLINTEC, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Björn Önfelt
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden.
- Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
8
|
Qin Y, Mace EM, Barton JP. An inference model gives insights into innate immune adaptation and repertoire diversity. Proc Natl Acad Sci U S A 2023; 120:e2305859120. [PMID: 37695895 PMCID: PMC10515141 DOI: 10.1073/pnas.2305859120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 08/08/2023] [Indexed: 09/13/2023] Open
Abstract
The innate immune system is the body's first line of defense against infection. Natural killer (NK) cells, a vital part of the innate immune system, help to control infection and eliminate cancer. Studies have identified a vast array of receptors that NK cells use to discriminate between healthy and unhealthy cells. However, at present, it is difficult to explain how NK cells will respond to novel stimuli in different environments. In addition, the expression of different receptors on individual NK cells is highly stochastic, but the reason for these variegated expression patterns is unclear. Here, we studied the recognition of unhealthy target cells as an inference problem, where NK cells must distinguish between healthy targets with normal variability in ligand expression and ones that are clear "outliers." Our mathematical model fits well with experimental data, including NK cells' adaptation to changing environments and responses to different target cells. Furthermore, we find that stochastic, "sparse" receptor expression profiles are best able to detect a variety of possible threats, in agreement with experimental studies of the NK cell repertoire. While our study was specifically motivated by NK cells, our model is general and could also apply more broadly to explain principles of target recognition for other immune cell types.
Collapse
Affiliation(s)
- Yawei Qin
- Department of Physics and Astronomy, University of California, Riverside, CA92521
| | - Emily M. Mace
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY10032
| | - John P. Barton
- Department of Physics and Astronomy, University of California, Riverside, CA92521
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA15260
| |
Collapse
|
9
|
Carannante V, Wiklund M, Önfelt B. In vitro models to study natural killer cell dynamics in the tumor microenvironment. Front Immunol 2023; 14:1135148. [PMID: 37457703 PMCID: PMC10338882 DOI: 10.3389/fimmu.2023.1135148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 06/05/2023] [Indexed: 07/18/2023] Open
Abstract
Immunotherapy is revolutionizing cancer therapy. The rapid development of new immunotherapeutic strategies to treat solid tumors is posing new challenges for preclinical research, demanding novel in vitro methods to test treatments. Such methods should meet specific requirements, such as enabling the evaluation of immune cell responses like cytotoxicity or cytokine release, and infiltration into the tumor microenvironment using cancer models representative of the original disease. They should allow high-throughput and high-content analysis, to evaluate the efficacy of treatments and understand immune-evasion processes to facilitate development of new therapeutic targets. Ideally, they should be suitable for personalized immunotherapy testing, providing information for patient stratification. Consequently, the application of in vitro 3-dimensional (3D) cell culture models, such as tumor spheroids and organoids, is rapidly expanding in the immunotherapeutic field, coupled with the development of novel imaging-based techniques and -omic analysis. In this paper, we review the recent advances in the development of in vitro 3D platforms applied to natural killer (NK) cell-based cancer immunotherapy studies, highlighting the benefits and limitations of the current methods, and discuss new concepts and future directions of the field.
Collapse
Affiliation(s)
- Valentina Carannante
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Martin Wiklund
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Björn Önfelt
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
- Center for Infectious Medicine, Department of Medicine Huddinge, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
10
|
Mace EM. Human natural killer cells: Form, function, and development. J Allergy Clin Immunol 2023; 151:371-385. [PMID: 36195172 PMCID: PMC9905317 DOI: 10.1016/j.jaci.2022.09.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 08/22/2022] [Accepted: 09/02/2022] [Indexed: 02/07/2023]
Abstract
Human natural killer (NK) cells are innate lymphoid cells that mediate important effector functions in the control of viral infection and malignancy. Their ability to distinguish "self" from "nonself" and lyse virally infected and tumorigenic cells through germline-encoded receptors makes them important players in maintaining human health and a powerful tool for immunotherapeutic applications and fighting disease. This review introduces our current understanding of NK cell biology, including key facets of NK cell differentiation and the acquisition and execution of NK cell effector function. Further, it addresses the clinical relevance of NK cells in both primary immunodeficiency and immunotherapy. It is intended to provide an up-to-date and comprehensive overview of this important and interesting innate immune effector cell subset.
Collapse
Affiliation(s)
- Emily M Mace
- Division of Allergy, Immunology and Rheumatology, Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York.
| |
Collapse
|
11
|
Li B, Ma X, Cheng J, Tian T, Guo J, Wang Y, Pang L. Droplets microfluidics platform-A tool for single cell research. Front Bioeng Biotechnol 2023; 11:1121870. [PMID: 37152651 PMCID: PMC10154550 DOI: 10.3389/fbioe.2023.1121870] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 04/03/2023] [Indexed: 05/09/2023] Open
Abstract
Cells are the most basic structural and functional units of living organisms. Studies of cell growth, differentiation, apoptosis, and cell-cell interactions can help scientists understand the mysteries of living systems. However, there is considerable heterogeneity among cells. Great differences between individuals can be found even within the same cell cluster. Cell heterogeneity can only be clearly expressed and distinguished at the level of single cells. The development of droplet microfluidics technology opens up a new chapter for single-cell analysis. Microfluidic chips can produce many nanoscale monodisperse droplets, which can be used as small isolated micro-laboratories for various high-throughput, precise single-cell analyses. Moreover, gel droplets with good biocompatibility can be used in single-cell cultures and coupled with biomolecules for various downstream analyses of cellular metabolites. The droplets are also maneuverable; through physical and chemical forces, droplets can be divided, fused, and sorted to realize single-cell screening and other related studies. This review describes the channel design, droplet generation, and control technology of droplet microfluidics and gives a detailed overview of the application of droplet microfluidics in single-cell culture, single-cell screening, single-cell detection, and other aspects. Moreover, we provide a recent review of the application of droplet microfluidics in tumor single-cell immunoassays, describe in detail the advantages of microfluidics in tumor research, and predict the development of droplet microfluidics at the single-cell level.
Collapse
Affiliation(s)
- Bixuan Li
- Xi’an Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi’an, China
- School of Basic Medicine, Xi’an Medical University, Xi’an, China
| | - Xi Ma
- Xi’an Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi’an, China
- School of Basic Medicine, Xi’an Medical University, Xi’an, China
| | - Jianghong Cheng
- Xi’an Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi’an, China
- School of Basic Medicine, Xi’an Medical University, Xi’an, China
| | - Tian Tian
- Xi’an Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi’an, China
- School of Basic Medicine, Xi’an Medical University, Xi’an, China
| | - Jiao Guo
- Xi’an Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi’an, China
- School of Basic Medicine, Xi’an Medical University, Xi’an, China
| | - Yang Wang
- Xi’an Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi’an, China
- School of Basic Medicine, Xi’an Medical University, Xi’an, China
- *Correspondence: Yang Wang,
| | - Long Pang
- Xi’an Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi’an, China
- School of Basic Medicine, Xi’an Medical University, Xi’an, China
| |
Collapse
|
12
|
Subedi N, Verhagen LP, de Jonge P, Van Eyndhoven LC, van Turnhout MC, Koomen V, Baudry J, Eyer K, Dolstra H, Tel J. Single‐Cell Profiling Reveals Functional Heterogeneity and Serial Killing in Human Peripheral and Ex Vivo‐Generated CD34+ Progenitor‐Derived Natural Killer Cells. Adv Biol (Weinh) 2022; 7:e2200207. [PMID: 36517083 DOI: 10.1002/adbi.202200207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 11/25/2022] [Indexed: 12/23/2022]
Abstract
Increasing evidence suggests that natural killer (NK) cells are composed of distinct functional subsets. This multifunctional role has made them an attractive choice for anticancer immunotherapy. A functional NK cell repertoire is generated through cellular education, resulting in a heterogeneous NK cell population with distinct capabilities responding to different stimuli. The application of a high-throughput droplet-based microfluidic platform allows monitoring of NK cell-target cell interactions at the single-cell level and in real-time. A variable response of single NK cells toward different target cells is observed, and a distinct population of NK cells (serial killers) capable of inducing multiple target lysis is identified. By assessing the cytotoxic dynamics, it is shown that single umbilical cord blood-derived CD34+ hematopoietic progenitor (HPC)-NK cells display superior antitumor cytotoxicity. With an integrated analysis of cytotoxicity and cytokine secretion, it is shown that target cell interactions augment cytotoxic as well as secretory behavior of NK cells. By providing an integrated assessment of NK cell functions by microfluidics, this study paves the way to further functionally characterize NK cells ultimately aimed to improve cancer immunotherapy.
Collapse
Affiliation(s)
- Nikita Subedi
- Laboratory of Immunoengineering Department of Biomedical Engineering Eindhoven University of Technology Groene Loper 5 Eindhoven 5600 MB The Netherlands
- Institute for Complex Molecular Systems Eindhoven University of Technology Groene Loper 5 Eindhoven 5600 MB The Netherlands
| | - Liesbeth Petronella Verhagen
- Laboratory of Immunoengineering Department of Biomedical Engineering Eindhoven University of Technology Groene Loper 5 Eindhoven 5600 MB The Netherlands
- Institute for Complex Molecular Systems Eindhoven University of Technology Groene Loper 5 Eindhoven 5600 MB The Netherlands
| | - Paul de Jonge
- Department of Laboratory Medicine – Laboratory of Hematology Radboud Institute of Molecular Life Sciences Radboud University Medical Center Nijmegen 6525 GA The Netherlands
| | - Laura C. Van Eyndhoven
- Laboratory of Immunoengineering Department of Biomedical Engineering Eindhoven University of Technology Groene Loper 5 Eindhoven 5600 MB The Netherlands
- Institute for Complex Molecular Systems Eindhoven University of Technology Groene Loper 5 Eindhoven 5600 MB The Netherlands
| | - Mark C. van Turnhout
- Soft Tissue Engineering and Mechanobiology Department of Biomedical Engineering Eindhoven University of Technology Groene Loper 5 Eindhoven 5600 MB The Netherlands
| | - Vera Koomen
- Laboratory of Immunoengineering Department of Biomedical Engineering Eindhoven University of Technology Groene Loper 5 Eindhoven 5600 MB The Netherlands
| | - Jean Baudry
- Laboratoire Colloïdes et Matériaux Divisés (LCMD) ESPCI Paris PSL Research University CNRS UMR8231 Chimie Biologie Innovation Paris 75005 France
| | - Klaus Eyer
- Laboratoire Colloïdes et Matériaux Divisés (LCMD) ESPCI Paris PSL Research University CNRS UMR8231 Chimie Biologie Innovation Paris 75005 France
- Laboratory for Functional Immune Repertoire Analysis Institute of Pharmaceutical Sciences D‐CHAB, ETH, Zürich Zurich 8093 Switzerland
| | - Harry Dolstra
- Department of Laboratory Medicine – Laboratory of Hematology Radboud Institute of Molecular Life Sciences Radboud University Medical Center Nijmegen 6525 GA The Netherlands
| | - Jurjen Tel
- Laboratory of Immunoengineering Department of Biomedical Engineering Eindhoven University of Technology Groene Loper 5 Eindhoven 5600 MB The Netherlands
- Institute for Complex Molecular Systems Eindhoven University of Technology Groene Loper 5 Eindhoven 5600 MB The Netherlands
| |
Collapse
|
13
|
Haroun-Izquierdo A, Vincenti M, Netskar H, van Ooijen H, Zhang B, Bendzick L, Kanaya M, Momayyezi P, Li S, Wiiger MT, Hoel HJ, Krokeide SZ, Kremer V, Tjonnfjord G, Berggren S, Wikström K, Blomberg P, Alici E, Felices M, Önfelt B, Höglund P, Valamehr B, Ljunggren HG, Björklund A, Hammer Q, Kveberg L, Cichocki F, Miller JS, Malmberg KJ, Sohlberg E. Adaptive single-KIR +NKG2C + NK cells expanded from select superdonors show potent missing-self reactivity and efficiently control HLA-mismatched acute myeloid leukemia. J Immunother Cancer 2022; 10:jitc-2022-005577. [PMID: 36319065 PMCID: PMC9628692 DOI: 10.1136/jitc-2022-005577] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Natural killer (NK) cells hold great promise as a source for allogeneic cell therapy against hematological malignancies, including acute myeloid leukemia (AML). Current treatments are hampered by variability in NK cell subset responses, a limitation which could be circumvented by specific expansion of highly potent single killer immunoglobulin-like receptor (KIR)+NKG2C+ adaptive NK cells to maximize missing-self reactivity. METHODS We developed a GMP-compliant protocol to expand adaptive NK cells from cryopreserved cells derived from select third-party superdonors, that is, donors harboring large adaptive NK cell subsets with desired KIR specificities at baseline. We studied the adaptive state of the cell product (ADAPT-NK) by flow cytometry and mass cytometry as well as cellular indexing of transcriptomes and epitopes by sequencing (CITE-Seq). We investigated the functional responses of ADAPT-NK cells against a wide range of tumor target cell lines and primary AML samples using flow cytometry and IncuCyte as well as in a mouse model of AML. RESULTS ADAPT-NK cells were >90% pure with a homogeneous expression of a single self-HLA specific KIR and expanded a median of 470-fold. The ADAPT-NK cells largely retained their adaptive transcriptional signature with activation of effector programs without signs of exhaustion. ADAPT-NK cells showed high degranulation capacity and efficient killing of HLA-C/KIR mismatched tumor cell lines as well as primary leukemic blasts from AML patients. Finally, the expanded adaptive NK cells had preserved robust antibody-dependent cellular cytotoxicity potential and combination of ADAPT-NK cells with an anti-CD16/IL-15/anti-CD33 tri-specific engager led to near-complete killing of resistant CD45dim blast subtypes. CONCLUSIONS These preclinical data demonstrate the feasibility of off-the-shelf therapy with a non-engineered, yet highly specific, NK cell population with full missing-self recognition capability.
Collapse
Affiliation(s)
- Alvaro Haroun-Izquierdo
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Marianna Vincenti
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Herman Netskar
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Hanna van Ooijen
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Bin Zhang
- University of Minnesota, Masonic Cancer Center, Minneapolis, Minnesota, USA
| | - Laura Bendzick
- University of Minnesota, Masonic Cancer Center, Minneapolis, Minnesota, USA
| | - Minoru Kanaya
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Pouria Momayyezi
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Shuo Li
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Merete Thune Wiiger
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Hanna Julie Hoel
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Silje Zandstra Krokeide
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Veronika Kremer
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Geir Tjonnfjord
- Department of Hematology, Oslo University Hospital and K.G. Jebsen Centre for B-cell malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Stéphanie Berggren
- Vecura, Karolinska Center for Cell Therapy Clinical Research Center, Karolinska University Hospital, Stockholm, Sweden
| | - Kristina Wikström
- Vecura, Karolinska Center for Cell Therapy Clinical Research Center, Karolinska University Hospital, Stockholm, Sweden
| | - Pontus Blomberg
- Vecura, Karolinska Center for Cell Therapy Clinical Research Center, Karolinska University Hospital, Stockholm, Sweden
| | - Evren Alici
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Martin Felices
- University of Minnesota, Masonic Cancer Center, Minneapolis, Minnesota, USA
| | - Björn Önfelt
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Petter Höglund
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | | | - Hans-Gustaf Ljunggren
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Andreas Björklund
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation, Karolinska University Hospital, Stockholm, Sweden
| | - Quirin Hammer
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Lise Kveberg
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Frank Cichocki
- University of Minnesota, Masonic Cancer Center, Minneapolis, Minnesota, USA
| | - Jeffrey S Miller
- University of Minnesota, Masonic Cancer Center, Minneapolis, Minnesota, USA
| | - Karl-Johan Malmberg
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Ebba Sohlberg
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
14
|
Kim SE, Yun S, Doh J. Effects of extracellular adhesion molecules on immune cell mediated solid tumor cell killing. Front Immunol 2022; 13:1004171. [PMID: 36389663 PMCID: PMC9647090 DOI: 10.3389/fimmu.2022.1004171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 10/10/2022] [Indexed: 01/27/2023] Open
Abstract
Adoptive cell therapy (ACT) using ex vivo engineered/expanded immune cells demonstrated poor efficacy against solid tumors, despite its great success in treating various hematopoietic malignancies. To improve ACT for solid tumors, it is crucial to comprehend how the numerous components of the tumor microenvironment (TME) surrounding solid tumor cells influence killing ability of immune cells. In this study, we sought to determine the effects of extracellular adhesion provided by extracellular matrix (ECM) of TME on immune cell cytotoxicity by devising microwell arrays coated with proteins either preventing or promoting cell adhesion. Solid tumor cells in bovine serum albumin (BSA)-coated microwells did not attach to the surfaces and exhibited a round morphology, but solid tumor cells in fibronectin (FN)-coated microwells adhered firmed to the substrates with a flat shape. The seeding densities of solid tumor cells and immune cells were tuned to maximize one-to-one pairing within a single microwell, and live cell imaging was performed to examine dynamic cell-cell interactions and immune cell cytotoxicity at a single cell level. Both natural killer (NK) cells and T cells showed higher cytotoxicity against round tumor cells in BSA-coated microwells compared to flat tumor cells in FN-coated microwells, suggesting that extracellular adhesion-mediated firm adhesion of tumor cells made them more resistant to immune cell-mediated killing. Additionally, NK cells and T cells in FN-coated microwells exhibited divergent dynamic behaviors, indicating that two distinct subsets of cytotoxic lymphocytes respond differentially to extracellular adhesion cues during target cell recognition.
Collapse
Affiliation(s)
- Seong-Eun Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, South Korea
| | - Suji Yun
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, South Korea
| | - Junsang Doh
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, South Korea,Department of Materials Science and Engineering, Research Institute of Advanced Materials, Institute of Engineering Research, Bio-MAX Institute, Soft Foundry Institute, Seoul National University, Seoul, South Korea,*Correspondence: Junsang Doh,
| |
Collapse
|
15
|
Sandström N, Carannante V, Olofsson K, Sandoz PA, Moussaud-Lamodière EL, Seashore-Ludlow B, Van Ooijen H, Verron Q, Frisk T, Takai M, Wiklund M, Östling P, Önfelt B. Miniaturized and multiplexed high-content screening of drug and immune sensitivity in a multichambered microwell chip. CELL REPORTS METHODS 2022; 2:100256. [PMID: 35880015 PMCID: PMC9308168 DOI: 10.1016/j.crmeth.2022.100256] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 04/21/2022] [Accepted: 06/17/2022] [Indexed: 12/01/2022]
Abstract
Here, we present a methodology based on multiplexed fluorescence screening of two- or three-dimensional cell cultures in a newly designed multichambered microwell chip, allowing direct assessment of drug or immune cell cytotoxic efficacy. We establish a framework for cell culture, formation of tumor spheroids, fluorescence labeling, and imaging of fixed or live cells at various magnifications directly in the chip together with data analysis and interpretation. The methodology is demonstrated by drug cytotoxicity screening using ovarian and non-small cell lung cancer cells and by cellular cytotoxicity screening targeting tumor spheroids of renal carcinoma and ovarian carcinoma with natural killer cells from healthy donors. The miniaturized format allowing long-term cell culture, efficient screening, and high-quality imaging of small sample volumes makes this methodology promising for individualized cytotoxicity tests for precision medicine.
Collapse
Affiliation(s)
- Niklas Sandström
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, 171 65 Solna, Sweden
| | - Valentina Carannante
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, 171 65 Solna, Sweden
- Department of Microbiology, Tumor and Cell Biology, Science for Life Laboratory, Karolinska Institutet, 171 65 Solna, Sweden
| | - Karl Olofsson
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, 171 65 Solna, Sweden
| | - Patrick A. Sandoz
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, 171 65 Solna, Sweden
| | | | - Brinton Seashore-Ludlow
- Department of Oncology and Pathology, Science for Life Laboratory, Karolinska Institutet, 171 65 Solna, Sweden
| | - Hanna Van Ooijen
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, 171 65 Solna, Sweden
| | - Quentin Verron
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, 171 65 Solna, Sweden
| | - Thomas Frisk
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, 171 65 Solna, Sweden
| | - Madoka Takai
- Department of Bioengineering, University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo 113-8656, Japan
| | - Martin Wiklund
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, 171 65 Solna, Sweden
| | - Päivi Östling
- Department of Oncology and Pathology, Science for Life Laboratory, Karolinska Institutet, 171 65 Solna, Sweden
| | - Björn Önfelt
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, 171 65 Solna, Sweden
- Department of Microbiology, Tumor and Cell Biology, Science for Life Laboratory, Karolinska Institutet, 171 65 Solna, Sweden
| |
Collapse
|
16
|
Tiemeijer BM, Tel J. Hydrogels for Single-Cell Microgel Production: Recent Advances and Applications. Front Bioeng Biotechnol 2022; 10:891461. [PMID: 35782502 PMCID: PMC9247248 DOI: 10.3389/fbioe.2022.891461] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 05/09/2022] [Indexed: 12/12/2022] Open
Abstract
Single-cell techniques have become more and more incorporated in cell biological research over the past decades. Various approaches have been proposed to isolate, culture, sort, and analyze individual cells to understand cellular heterogeneity, which is at the foundation of every systematic cellular response in the human body. Microfluidics is undoubtedly the most suitable method of manipulating cells, due to its small scale, high degree of control, and gentle nature toward vulnerable cells. More specifically, the technique of microfluidic droplet production has proven to provide reproducible single-cell encapsulation with high throughput. Various in-droplet applications have been explored, ranging from immunoassays, cytotoxicity assays, and single-cell sequencing. All rely on the theoretically unlimited throughput that can be achieved and the monodispersity of each individual droplet. To make these platforms more suitable for adherent cells or to maintain spatial control after de-emulsification, hydrogels can be included during droplet production to obtain “microgels.” Over the past years, a multitude of research has focused on the possibilities these can provide. Also, as the technique matures, it is becoming clear that it will result in advantages over conventional droplet approaches. In this review, we provide a comprehensive overview on how various types of hydrogels can be incorporated into different droplet-based approaches and provide novel and more robust analytic and screening applications. We will further focus on a wide range of recently published applications for microgels and how these can be applied in cell biological research at the single- to multicell scale.
Collapse
Affiliation(s)
- B. M. Tiemeijer
- Laboratory of Immunoengineering, Department of Biomedical Engineering, TU Eindhoven, Eindhoven, Netherlands
- Institute of Complex Molecular Systems, TU Eindhoven, Eindhoven, Netherlands
| | - J. Tel
- Laboratory of Immunoengineering, Department of Biomedical Engineering, TU Eindhoven, Eindhoven, Netherlands
- Institute of Complex Molecular Systems, TU Eindhoven, Eindhoven, Netherlands
- *Correspondence: J. Tel,
| |
Collapse
|
17
|
Sandström N, Brandt L, Sandoz PA, Zambarda C, Guldevall K, Schulz-Ruhtenberg M, Rösener B, Krüger RA, Önfelt B. Live single cell imaging assays in glass microwells produced by laser-induced deep etching. LAB ON A CHIP 2022; 22:2107-2121. [PMID: 35470832 DOI: 10.1039/d2lc00090c] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Miniaturization of cell culture substrates enables controlled analysis of living cells in confined micro-scale environments. This is particularly suitable for imaging individual cells over time, as they can be monitored without escaping the imaging field-of-view (FoV). Glass materials are ideal for most microscopy applications. However, with current methods used in life sciences, glass microfabrication is limited in terms of either freedom of design, quality, or throughput. In this work, we introduce laser-induced deep etching (LIDE) as a method for producing glass microwell arrays for live single cell imaging assays. We demonstrate novel microwell arrays with deep, high-aspect ratio wells that have rounded, dimpled or flat bottom profiles in either single-layer or double-layer glass chips. The microwells are evaluated for microscopy-based analysis of long-term cell culture, clonal expansion, laterally organized cell seeding, subcellular mechanics during migration and immune cell cytotoxicity assays of both adherent and suspension cells. It is shown that all types of microwells can support viable cell cultures and imaging with single cell resolution, and we highlight specific benefits of each microwell design for different applications. We believe that high-quality glass microwell arrays enabled by LIDE provide a great option for high-content and high-resolution imaging-based live cell assays with a broad range of potential applications within life sciences.
Collapse
Affiliation(s)
- Niklas Sandström
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden.
| | - Ludwig Brandt
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden.
| | - Patrick A Sandoz
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden.
| | - Chiara Zambarda
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden.
| | - Karolin Guldevall
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden.
| | | | | | | | - Björn Önfelt
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden.
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
18
|
Navarrete-Galvan L, Guglielmo M, Cruz Amaya J, Smith-Gagen J, Lombardi VC, Merica R, Hudig D. Optimizing NK-92 serial killers: gamma irradiation, CD95/Fas-ligation, and NK or LAK attack limit cytotoxic efficacy. J Transl Med 2022; 20:151. [PMID: 35366943 PMCID: PMC8976335 DOI: 10.1186/s12967-022-03350-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/15/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The NK cell line NK-92 and its genetically modified variants are receiving attention as immunotherapies to treat a range of malignancies. However, since NK-92 cells are themselves tumors, they require irradiation prior to transfer and are potentially susceptible to attack by patients' immune systems. Here, we investigated NK-92 cell-mediated serial killing for the effects of gamma-irradiation and ligation of the death receptor Fas (CD95), and NK-92 cell susceptibility to attack by activated primary blood NK cells. METHODS To evaluate serial killing, we used 51Cr-release assays with low NK-92 effector cell to target Raji, Daudi or K562 tumor cell (E:T) ratios to determine killing frequencies at 2-, 4-, 6-, and 8-h. RESULTS NK-92 cells were able to kill up to 14 Raji cells per NK-92 cell in 8 h. NK-92 cells retained high cytotoxic activity immediately after irradiation with 10 Gy but the cells surviving irradiation lost > 50% activity 1 day after irradiation. Despite high expression of CD95, NK-92 cells maintained their viability following overnight Fas/CD95-ligation but lost some cytotoxic activity. However, 1 day after irradiation, NK-92 cells were more susceptible to Fas ligation, resulting in decreased cytotoxic activity of the cells surviving irradiation. Irradiated NK-92 cells were also susceptible to killing by both unstimulated and IL-2 activated primary NK cells (LAK). In contrast, non-irradiated NK-92 cells were more resistant to attack by NK and LAK cells. CONCLUSIONS Irradiation is deleterious to both the survival and cytotoxicity mediated by NK-92 cells and renders the NK-92 cells susceptible to Fas-initiated death and death initiated by primary blood NK cells. Therefore, replacement of irradiation as an antiproliferative pretreatment and genetic deletion of Fas and/or NK activation ligands from adoptively transferred cell lines are indicated as new approaches to increase therapeutic efficacy.
Collapse
Affiliation(s)
| | | | | | - Julie Smith-Gagen
- University of Nevada, Reno School of Community Health Sciences, Reno, NV, 89557, USA
| | | | - Rebecca Merica
- Biology Department, St. Olaf College, Northfield, MN, 55057, USA
| | - Dorothy Hudig
- University of Nevada, Reno School of Medicine, Reno, NV, 89557, USA.
| |
Collapse
|
19
|
Ben-Shmuel A, Sabag B, Puthenveetil A, Biber G, Levy M, Jubany T, Awwad F, Roy RK, Joseph N, Matalon O, Kivelevitz J, Barda-Saad M. Inhibition of SHP-1 activity by PKC-θ regulates NK cell activation threshold and cytotoxicity. eLife 2022; 11:73282. [PMID: 35258455 PMCID: PMC8903836 DOI: 10.7554/elife.73282] [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: 08/23/2021] [Accepted: 02/23/2022] [Indexed: 12/26/2022] Open
Abstract
Natural killer (NK) cells play a crucial role in immunity, killing virally infected and cancerous cells. The balance of signals initiated upon engagement of activating and inhibitory NK receptors with cognate ligands determines killing or tolerance. Nevertheless, the molecular mechanisms regulating rapid NK cell discrimination between healthy and malignant cells in a heterogeneous tissue environment are incompletely understood. The SHP-1 tyrosine phosphatase is the central negative NK cell regulator that dephosphorylates key activating signaling proteins. Though the mechanism by which SHP-1 mediates NK cell inhibition has been partially elucidated, the pathways by which SHP-1 is itself regulated remain unclear. Here, we show that phosphorylation of SHP-1 in NK cells on the S591 residue by PKC-θ promotes the inhibited SHP-1 ‘folded’ state. Silencing PKC-θ maintains SHP-1 in the active conformation, reduces NK cell activation and cytotoxicity, and promotes tumor progression in vivo. This study reveals a molecular pathway that sustains the NK cell activation threshold through suppression of SHP-1 activity.
Collapse
Affiliation(s)
- Aviad Ben-Shmuel
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Batel Sabag
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Abhishek Puthenveetil
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Guy Biber
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Moria Levy
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Tammir Jubany
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Fatima Awwad
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Roshan Kumar Roy
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Noah Joseph
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Omri Matalon
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Jessica Kivelevitz
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Mira Barda-Saad
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| |
Collapse
|
20
|
Understanding natural killer cell biology from a single cell perspective. Cell Immunol 2022; 373:104497. [DOI: 10.1016/j.cellimm.2022.104497] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/26/2022] [Accepted: 02/16/2022] [Indexed: 12/27/2022]
|
21
|
Yoon J, Kang Y, Kim H, Torati SR, Kim K, Lim B, Kim C. Magnetophoretic Micro-Distributor for Controlled Clustering of Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103579. [PMID: 34910376 PMCID: PMC8867205 DOI: 10.1002/advs.202103579] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/31/2021] [Indexed: 06/14/2023]
Abstract
Cell clustering techniques are important to produce artificial cell clusters for in vitro models of intercellular mechanisms at the single-cell level. The analyses considering physical variables such as the shape and size of cells have been very limited. In addition, the precise manipulation of cells and control of the physical variables are still challenging. In this paper, a magnetophoretic device consisting of a trampoline micromagnet and active elements that enable the control of individual selective jumping motion and positioning of a micro-object is proposed. Based on a numerical simulation under various conditions, automatic separation or selective clustering of micro-objects according to their sizes is performed by parallel control and programmable manipulation. This method provides efficient control of the physical variables of cells and grouping of cells with the desired size and number, which can be a milestone for a better understanding of the intercellular dynamics between clustered cells at the single-cell level for future cell-on-chip applications.
Collapse
Affiliation(s)
- Jonghwan Yoon
- Department of Emerging Materials ScienceDGISTDaegu42988Republic of Korea
| | - Yumin Kang
- Department of Emerging Materials ScienceDGISTDaegu42988Republic of Korea
| | - Hyeonseol Kim
- Department of Emerging Materials ScienceDGISTDaegu42988Republic of Korea
| | - Sri Ramulu Torati
- Department of Emerging Materials ScienceDGISTDaegu42988Republic of Korea
| | - Keonmok Kim
- Department of Emerging Materials ScienceDGISTDaegu42988Republic of Korea
| | - Byeonghwa Lim
- Department of Emerging Materials ScienceDGISTDaegu42988Republic of Korea
| | - CheolGi Kim
- Department of Emerging Materials ScienceDGISTDaegu42988Republic of Korea
| |
Collapse
|
22
|
Tuomela K, Mukherjee D, Ambrose AR, Harikrishnan A, Mole H, Hurlstone A, Önfelt B, Honeychurch J, Davis DM. Radiotherapy transiently reduces the sensitivity of cancer cells to lymphocyte cytotoxicity. Proc Natl Acad Sci U S A 2022; 119:e2111900119. [PMID: 35042775 PMCID: PMC8785960 DOI: 10.1073/pnas.2111900119] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 12/07/2021] [Indexed: 12/13/2022] Open
Abstract
The impact of radiotherapy on the interaction between immune cells and cancer cells is important not least because radiotherapy can be used alongside immunotherapy as a cancer treatment. Unexpectedly, we found that X-ray irradiation of cancer cells induced significant resistance to natural killer (NK) cell killing. This was true across a wide variety of cancer-cell types as well as for antibody-dependent cellular cytotoxicity. Resistance appeared 72 h postirradiation and persisted for 2 wk. Resistance could also occur independently of radiotherapy through pharmacologically induced cell-cycle arrest. Crucially, multiple steps in NK-cell engagement, synapse assembly, and activation were unaffected by target cell irradiation. Instead, radiotherapy caused profound resistance to perforin-induced calcium flux and lysis. Resistance also occurred to a structurally similar bacterial toxin, streptolysin O. Radiotherapy did not affect the binding of pore-forming proteins at the cell surface or membrane repair. Rather, irradiation instigated a defect in functional pore formation, consistent with phosphatidylserine-mediated perforin inhibition. In vivo, radiotherapy also led to a significant reduction in NK cell-mediated clearance of cancer cells. Radiotherapy-induced resistance to perforin also constrained chimeric antigen receptor T-cell cytotoxicity. Together, these data establish a treatment-induced resistance to lymphocyte cytotoxicity that is important to consider in the design of radiotherapy-immunotherapy protocols.
Collapse
Affiliation(s)
- Karoliina Tuomela
- The Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester M13 9NT, United Kingdom
| | - Debayan Mukherjee
- Division of Cancer Sciences, The University of Manchester, Manchester M13 9PL, United Kingdom
| | - Ashley R Ambrose
- The Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester M13 9NT, United Kingdom
| | - Ashish Harikrishnan
- The Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester M13 9NT, United Kingdom
| | - Holly Mole
- The Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester M13 9NT, United Kingdom
| | - Adam Hurlstone
- The Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester M13 9NT, United Kingdom
| | - Björn Önfelt
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden
- Department of Applied Physics, Science for Life Laboratory, Kungliga Tekniska Högskolan Royal Institute of Technology, 17165 Stockholm, Sweden
| | - Jamie Honeychurch
- Division of Cancer Sciences, The University of Manchester, Manchester M13 9PL, United Kingdom
| | - Daniel M Davis
- The Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester M13 9NT, United Kingdom;
| |
Collapse
|
23
|
An automated real-time microfluidic platform to probe single NK cell heterogeneity and cytotoxicity on-chip. Sci Rep 2021; 11:17084. [PMID: 34429486 PMCID: PMC8385055 DOI: 10.1038/s41598-021-96609-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 08/03/2021] [Indexed: 12/28/2022] Open
Abstract
Cytotoxicity is a vital effector mechanism used by immune cells to combat pathogens and cancer cells. While conventional cytotoxicity assays rely on averaged end-point measures, crucial insights on the dynamics and heterogeneity of effector and target cell interactions cannot be extracted, emphasizing the need for dynamic single-cell analysis. Here, we present a fully automated droplet-based microfluidic platform that allowed the real-time monitoring of effector-target cell interactions and killing, allowing the screening of over 60,000 droplets identifying 2000 individual cellular interactions monitored over 10 h. During the course of incubation, we observed that the dynamics of cytotoxicity within the Natural Killer (NK) cell population varies significantly over the time. Around 20% of the total NK cells in droplets showed positive cytotoxicity against paired K562 cells, most of which was exhibited within first 4 h of cellular interaction. Using our single cell analysis platform, we demonstrated that the population of NK cells is composed of individual cells with different strength in their effector functions, a behavior masked in conventional studies. Moreover, the versatility of our platform will allow the dynamic and resolved study of interactions between immune cell types and the finding and characterization of functional sub-populations, opening novel ways towards both fundamental and translational research.
Collapse
|
24
|
Bhingardive V, Le Saux G, Edri A, Porgador A, Schvartzman M. Nanowire Based Guidance of the Morphology and Cytotoxic Activity of Natural Killer Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2007347. [PMID: 33719212 DOI: 10.1002/smll.202007347] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 01/13/2021] [Indexed: 06/12/2023]
Abstract
The cytotoxic activity of natural killer (NK) cells is regulated by many chemical and physical cues, whose integration mechanism is still obscure. Here, a multifunctional platform is engineered for NK cell stimulation, to study the effect of the signal integration and spatial heterogeneity on NK cell function. The platform is based on nanowires, whose mechanical compliance and site-selective tip functionalization with antigens produce the physical and chemical stimuli, respectively. The nanowires are confined to micron-sized islands, which induce a splitting of the NK cells into two subpopulations with distinct morphologies and immune responses: NK cells atop the nanowire islands display symmetrical spreading and enhanced activation, whereas cells lying in the straits between the islands develop elongated profiles and show lower activation levels. The demonstrated tunability of NK cell cytotoxicity provides an important insight into the mechanism of their immune function and introduces a novel technological route for the ex vivo shaping of cytotoxic lymphocytes in immunotherapy.
Collapse
Affiliation(s)
- Viraj Bhingardive
- Department of Materials Engineering, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva, 84105, Israel
- Ilse Katz Institute for Nanoscale Science and Technology, The Shraga Segal Department of Microbiology, Immunology, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva, 84105, Israel
| | - Guillaume Le Saux
- Department of Materials Engineering, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva, 84105, Israel
- Ilse Katz Institute for Nanoscale Science and Technology, The Shraga Segal Department of Microbiology, Immunology, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva, 84105, Israel
| | - Avishay Edri
- Genetics Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva, 84105, Israel
| | - Angel Porgador
- Genetics Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva, 84105, Israel
| | - Mark Schvartzman
- Department of Materials Engineering, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva, 84105, Israel
- Ilse Katz Institute for Nanoscale Science and Technology, The Shraga Segal Department of Microbiology, Immunology, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva, 84105, Israel
| |
Collapse
|
25
|
Lafouresse F, Jugele R, Müller S, Doineau M, Duplan-Eche V, Espinosa E, Puisségur MP, Gadat S, Valitutti S. Stochastic asymmetric repartition of lytic machinery in dividing CD8 + T cells generates heterogeneous killing behavior. eLife 2021; 10:62691. [PMID: 33427199 PMCID: PMC7867409 DOI: 10.7554/elife.62691] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 01/08/2021] [Indexed: 12/27/2022] Open
Abstract
Cytotoxic immune cells are endowed with a high degree of heterogeneity in their lytic function, but how this heterogeneity is generated is still an open question. We therefore investigated if human CD8+ T cells could segregate their lytic components during telophase, using imaging flow cytometry, confocal microscopy, and live-cell imaging. We show that CD107a+-intracellular vesicles, perforin, and granzyme B unevenly segregate in a constant fraction of telophasic cells during each division round. Mathematical modeling posits that unequal lytic molecule inheritance by daughter cells results from the random distribution of lytic granules on the two sides of the cleavage furrow. Finally, we establish that the level of lytic compartment in individual cytotoxic T lymphocyte (CTL) dictates CTL killing capacity.
Collapse
Affiliation(s)
- Fanny Lafouresse
- INSERM U1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Université de Toulouse III-Paul Sabatier, Toulouse, France
| | - Romain Jugele
- INSERM U1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Université de Toulouse III-Paul Sabatier, Toulouse, France
| | - Sabina Müller
- INSERM U1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Université de Toulouse III-Paul Sabatier, Toulouse, France
| | - Marine Doineau
- Toulouse School of Economics, CNRS UMR 5314, Université Toulouse 1 Capitole, France and Institut Universitaire de France, Toulouse, France
| | - Valérie Duplan-Eche
- INSERM, UMR1043, Centre de Physiopathologie de Toulouse Purpan, Toulouse, France
| | - Eric Espinosa
- INSERM U1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Université de Toulouse III-Paul Sabatier, Toulouse, France
| | - Marie-Pierre Puisségur
- INSERM U1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Université de Toulouse III-Paul Sabatier, Toulouse, France
| | - Sébastien Gadat
- Toulouse School of Economics, CNRS UMR 5314, Université Toulouse 1 Capitole, France and Institut Universitaire de France, Toulouse, France
| | - Salvatore Valitutti
- INSERM U1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Université de Toulouse III-Paul Sabatier, Toulouse, France.,Department of Pathology, Institut Universitaire du Cancer-Oncopole de Toulouse, Toulouse, France
| |
Collapse
|
26
|
Mordechay L, Le Saux G, Edri A, Hadad U, Porgador A, Schvartzman M. Mechanical Regulation of the Cytotoxic Activity of Natural Killer Cells. ACS Biomater Sci Eng 2020; 7:122-132. [DOI: 10.1021/acsbiomaterials.0c01121] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Lital Mordechay
- Department of Materials Engineering, Ben-Gurion University of the Negev, 84105 Beer Sheva, Israel
- Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev, 84105 Beer Sheva, Israel
| | - Guillaume Le Saux
- Department of Materials Engineering, Ben-Gurion University of the Negev, 84105 Beer Sheva, Israel
- Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev, 84105 Beer Sheva, Israel
| | - Avishay Edri
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, 84105 Beer Sheva, Israel
| | - Uzi Hadad
- Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev, 84105 Beer Sheva, Israel
| | - Angel Porgador
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, 84105 Beer Sheva, Israel
| | - Mark Schvartzman
- Department of Materials Engineering, Ben-Gurion University of the Negev, 84105 Beer Sheva, Israel
- Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev, 84105 Beer Sheva, Israel
| |
Collapse
|
27
|
Van der Meer JMR, Maas RJA, Guldevall K, Klarenaar K, de Jonge PKJD, Evert JSHV, van der Waart AB, Cany J, Safrit JT, Lee JH, Wagena E, Friedl P, Önfelt B, Massuger LF, Schaap NPM, Jansen JH, Hobo W, Dolstra H. IL-15 superagonist N-803 improves IFNγ production and killing of leukemia and ovarian cancer cells by CD34 + progenitor-derived NK cells. Cancer Immunol Immunother 2020; 70:1305-1321. [PMID: 33140189 PMCID: PMC8053152 DOI: 10.1007/s00262-020-02749-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 10/14/2020] [Indexed: 12/17/2022]
Abstract
Allogeneic natural killer (NK) cell transfer is a potential immunotherapy to eliminate and control cancer. A promising source are CD34 + hematopoietic progenitor cells (HPCs), since large numbers of cytotoxic NK cells can be generated. Effective boosting of NK cell function can be achieved by interleukin (IL)-15. However, its in vivo half-life is short and potent trans-presentation by IL-15 receptor α (IL-15Rα) is absent. Therefore, ImmunityBio developed IL-15 superagonist N-803, which combines IL-15 with an activating mutation, an IL-15Rα sushi domain for trans-presentation, and IgG1-Fc for increased half-life. Here, we investigated whether and how N-803 improves HPC-NK cell functionality in leukemia and ovarian cancer (OC) models in vitro and in vivo in OC-bearing immunodeficient mice. We used flow cytometry-based assays, enzyme-linked immunosorbent assay, microscopy-based serial killing assays, and bioluminescence imaging, for in vitro and in vivo experiments. N-803 increased HPC-NK cell proliferation and interferon (IFN)γ production. On leukemia cells, co-culture with HPC-NK cells and N-803 increased ICAM-1 expression. Furthermore, N-803 improved HPC-NK cell-mediated (serial) leukemia killing. Treating OC spheroids with HPC-NK cells and N-803 increased IFNγ-induced CXCL10 secretion, and target killing after prolonged exposure. In immunodeficient mice bearing human OC, N-803 supported HPC-NK cell persistence in combination with total human immunoglobulins to prevent Fc-mediated HPC-NK cell depletion. Moreover, this combination treatment decreased tumor growth. In conclusion, N-803 is a promising IL-15-based compound that boosts HPC-NK cell expansion and functionality in vitro and in vivo. Adding N-803 to HPC-NK cell therapy could improve cancer immunotherapy.
Collapse
Affiliation(s)
- J M R Van der Meer
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 8, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - R J A Maas
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 8, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - K Guldevall
- Department of Applied Physics, Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - K Klarenaar
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 8, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - P K J D de Jonge
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 8, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - J S Hoogstad-van Evert
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 8, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
- Department of Obstetrics and Gynecology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - A B van der Waart
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 8, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - J Cany
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 8, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | | | - J H Lee
- ImmunityBio, Culver City, CA, USA
| | - E Wagena
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - P Friedl
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
- David H. Koch Center for Applied Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Cancer Genomics Center, Utrecht, The Netherlands
| | - B Önfelt
- Department of Applied Physics, Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - L F Massuger
- Department of Obstetrics and Gynecology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - N P M Schaap
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - J H Jansen
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 8, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - W Hobo
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 8, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - H Dolstra
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 8, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
28
|
Tang JJJ, Sung AP, Guglielmo MJ, Navarrete-Galvan L, Redelman D, Smith-Gagen J, Hudig D. Natural Killer (NK) Cell Expression of CD2 as a Predictor of Serial Antibody-Dependent Cell-Mediated Cytotoxicity (ADCC). Antibodies (Basel) 2020; 9:antib9040054. [PMID: 33081115 PMCID: PMC7709134 DOI: 10.3390/antib9040054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/17/2020] [Accepted: 10/13/2020] [Indexed: 02/06/2023] Open
Abstract
NK cell ADCC supports monoclonal antibody anti-tumor therapies. We investigated serial ADCC and whether it could be predicted by NK phenotypes, including expression of CD16A, CD2 and perforin. CD16A, the NK receptor for antibodies, has AA158 valine or phenylalanine variants with different affinities for IgG. CD2, a costimulatory protein, associates with CD16A and can augment CD16A-signaling. Pore-forming perforin is essential for rapid NK-mediated killing. NK cells were monitored for their ADCC serial killing frequency (KF). KF is the average number of target cells killed per cell by a cytotoxic cell population. KF comparisons were made at 1:4 CD16pos NK effector:target ratios. ADCC was toward Daudi cells labeled with 51Cr and obinutuzumab anti-CD20 antibody. CD16A genotypes were determined by DNA sequencing. CD2, CD16A, and perforin expression was monitored by flow cytometry. Serial killing KFs varied two-fold among 24 donors and were independent of CD16A genotypes and perforin levels. However, high percentages of CD2pos of the CD16Apos NK cells and high levels of CD16A were associated with high KFs. ROC analysis indicated that the %CD2pos of CD16Apos NK cells can predict KFs. In conclusion, the extent of serial ADCC varies significantly among donors and appears predictable by the CD2posCD16Apos NK phenotype.
Collapse
Affiliation(s)
- Jennifer J.-J. Tang
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA; (J.J.-J.T.); (A.P.S.); (M.J.G.); (L.N.-G.)
| | - Alexander P. Sung
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA; (J.J.-J.T.); (A.P.S.); (M.J.G.); (L.N.-G.)
| | - Michael J. Guglielmo
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA; (J.J.-J.T.); (A.P.S.); (M.J.G.); (L.N.-G.)
| | - Lydia Navarrete-Galvan
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA; (J.J.-J.T.); (A.P.S.); (M.J.G.); (L.N.-G.)
| | - Doug Redelman
- Department of Physiology and Cell Biology, Reno School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA;
| | - Julie Smith-Gagen
- School of Community Health Sciences, University of Nevada, Reno, 1664 N. Virginia St., Reno, NV 89557, USA;
| | - Dorothy Hudig
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA; (J.J.-J.T.); (A.P.S.); (M.J.G.); (L.N.-G.)
- Correspondence: ; Tel.: +1-775-784-4430
| |
Collapse
|
29
|
Antona S, Platzman I, Spatz JP. Droplet-Based Cytotoxicity Assay: Implementation of Time-Efficient Screening of Antitumor Activity of Natural Killer Cells. ACS OMEGA 2020; 5:24674-24683. [PMID: 33015484 PMCID: PMC7528335 DOI: 10.1021/acsomega.0c03264] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 09/04/2020] [Indexed: 06/11/2023]
Abstract
Natural killer (NK) cells are key players of the innate immune system. Due to their rapid cytotoxicity against infectious pathogens, hematologic malignancies, and solid tumors, NK cells represent solid candidates for cell-based immunotherapy. Despite the progress made in recent years, the heterogeneity in their cytotoxic behavior represents a drawback. With the goal of screening the intrinsic diversity of NK cells, droplet-based microfluidic technology is exploited to develop a single-cell time-efficient cytotoxicity assay. Toward this end, NK-92 cells are coencapsulated with hematological tumor cell lines in water-in-oil droplets of different sizes and their cytotoxic activity is evaluated. The effect of droplet-based confinement on NK cytotoxicity is investigated by controlling the droplet volume. The successful optimization of the droplet size allows for time efficiency compared to cytotoxicity assays based on flow cytometry. Additionally, the ability of individual NK-92 cells to kill multiple target cells in series is explored, expanding the knowledge about the serial killing process dynamics. The developed droplet-based microfluidic assay does not require the labeling of NK cells and represents a step toward developing of a forthcoming process for the selection of NK cells with the highest cytotoxicity against specific targets.
Collapse
Affiliation(s)
- Silvia Antona
- Department
of Cellular Biophysics, Max Planck Institute
for Medical Research, Jahnstraße 29, 69120 Heidelberg, Germany
- Institute
for Molecular Systems Engineering (IMSE), Heidelberg University, Im Neuenheimer Feld 225, 69120 Heidelberg, Germany
| | - Ilia Platzman
- Department
of Cellular Biophysics, Max Planck Institute
for Medical Research, Jahnstraße 29, 69120 Heidelberg, Germany
- Institute
for Molecular Systems Engineering (IMSE), Heidelberg University, Im Neuenheimer Feld 225, 69120 Heidelberg, Germany
| | - Joachim P. Spatz
- Department
of Cellular Biophysics, Max Planck Institute
for Medical Research, Jahnstraße 29, 69120 Heidelberg, Germany
- Institute
for Molecular Systems Engineering (IMSE), Heidelberg University, Im Neuenheimer Feld 225, 69120 Heidelberg, Germany
- Max
Planck School Matter to Life, Jahnstraße 29, D-69120 Heidelberg, Germany
| |
Collapse
|
30
|
Dunst J, Glaros V, Englmaier L, Sandoz PA, Önfelt B, Kisielow J, Kreslavsky T. Recognition of synthetic polyanionic ligands underlies "spontaneous" reactivity of Vγ1 γδTCRs. J Leukoc Biol 2020; 107:1033-1044. [PMID: 31943366 PMCID: PMC7317387 DOI: 10.1002/jlb.2ma1219-392r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 12/05/2019] [Accepted: 12/11/2019] [Indexed: 01/02/2023] Open
Abstract
Although γδTCRs were discovered more than 30 yr ago, principles of antigen recognition by these receptors remain unclear and the nature of these antigens is largely elusive. Numerous studies reported that T cell hybridomas expressing several Vγ1-containing TCRs, including the Vγ1Vδ6 TCR of γδNKT cells, spontaneously secrete cytokines. This property was interpreted as recognition of a self-ligand expressed on the hybridoma cells themselves. Here, we revisited this finding using a recently developed reporter system and live single cell imaging. We confirmed strong spontaneous signaling by Vγ1Vδ6 and related TCRs, but not by TCRs from several other γδ or innate-like αβ T cells, and demonstrated that both γ and δ chains contributed to this reactivity. Unexpectedly, live single cell imaging showed that activation of this signaling did not require any interaction between cells. Further investigation revealed that the signaling is instead activated by interaction with negatively charged surfaces abundantly present under regular cell culture conditions and was abrogated when noncharged cell culture vessels were used. This mode of TCR signaling activation was not restricted to the reporter cell lines, as interaction with negatively charged surfaces also triggered TCR signaling in ex vivo Vγ1 γδ T cells. Taken together, these results explain long-standing observations on the spontaneous reactivity of Vγ1Vδ6 TCR and demonstrate an unexpected antigen presentation-independent mode of TCR activation by a spectrum of chemically unrelated polyanionic ligands.
Collapse
Affiliation(s)
- Josefine Dunst
- Department of Medicine, Division of Immunology and Allergy, Karolinska InstitutetKarolinska University HospitalStockholmSweden
- Center for Molecular MedicineKarolinska InstitutetStockholmSweden
| | - Vassilis Glaros
- Department of Medicine, Division of Immunology and Allergy, Karolinska InstitutetKarolinska University HospitalStockholmSweden
- Center for Molecular MedicineKarolinska InstitutetStockholmSweden
| | - Lukas Englmaier
- Department of Medicine, Division of Immunology and Allergy, Karolinska InstitutetKarolinska University HospitalStockholmSweden
- Center for Molecular MedicineKarolinska InstitutetStockholmSweden
| | - Patrick A. Sandoz
- Department of Applied PhysicsScience for Life LaboratoryKTH Royal Institute of TechnologyStockholmSweden
| | - Björn Önfelt
- Department of Applied PhysicsScience for Life LaboratoryKTH Royal Institute of TechnologyStockholmSweden
- Department of Microbiology, Tumor and Cell BiologyKarolinska InstituteSolnaSweden
| | - Jan Kisielow
- Institute of Molecular Health SciencesETHZurichSwitzerland
| | - Taras Kreslavsky
- Department of Medicine, Division of Immunology and Allergy, Karolinska InstitutetKarolinska University HospitalStockholmSweden
- Center for Molecular MedicineKarolinska InstitutetStockholmSweden
| |
Collapse
|
31
|
Khazen R, Müller S, Lafouresse F, Valitutti S, Cussat-Blanc S. Sequential adjustment of cytotoxic T lymphocyte densities improves efficacy in controlling tumor growth. Sci Rep 2019; 9:12308. [PMID: 31444380 PMCID: PMC6707257 DOI: 10.1038/s41598-019-48711-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 08/09/2019] [Indexed: 12/30/2022] Open
Abstract
Understanding the human cytotoxic T lymphocyte (CTL) biology is crucial to develop novel strategies aiming at maximizing their lytic capacity against cancer cells. Here we introduce an agent-based model, calibrated on population-scale experimental data that allows quantifying human CTL per capita killing. Our model highlights higher individual CTL killing capacity at lower CTL densities and fits experimental data of human melanoma cell killing. The model allows extending the analysis over prolonged time frames, difficult to investigate experimentally, and reveals that initial high CTL densities hamper efficacy to control melanoma growth. Computational analysis forecasts that sequential addition of fresh CTL cohorts improves tumor growth control. In vivo experimental data, obtained in a mouse melanoma model, confirm this prediction. Taken together, our results unveil the impact that sequential adjustment of cellular densities has on enhancing CTL efficacy over long-term confrontation with tumor cells. In perspective, they can be instrumental to refine CTL-based therapeutic strategies aiming at controlling tumor growth.
Collapse
Affiliation(s)
- Roxana Khazen
- Centre de Recherches en Cancérologie de Toulouse (CRCT), UMR 1037 INSERM/ Université Toulouse III Paul Sabatier, «Equipe labellisée Ligue Nationale contre le cancer 2018», INSERM, Toulouse, France.,INSERM U1223, Dynamics of Immune Responses Unit, Institut Pasteur, 75015, Paris, France
| | - Sabina Müller
- Centre de Recherches en Cancérologie de Toulouse (CRCT), UMR 1037 INSERM/ Université Toulouse III Paul Sabatier, «Equipe labellisée Ligue Nationale contre le cancer 2018», INSERM, Toulouse, France
| | - Fanny Lafouresse
- Centre de Recherches en Cancérologie de Toulouse (CRCT), UMR 1037 INSERM/ Université Toulouse III Paul Sabatier, «Equipe labellisée Ligue Nationale contre le cancer 2018», INSERM, Toulouse, France
| | - Salvatore Valitutti
- Centre de Recherches en Cancérologie de Toulouse (CRCT), UMR 1037 INSERM/ Université Toulouse III Paul Sabatier, «Equipe labellisée Ligue Nationale contre le cancer 2018», INSERM, Toulouse, France. .,Department of Pathology, Institut Universitaire du Cancer-Oncopole de Toulouse, 31059, Toulouse, France.
| | - Sylvain Cussat-Blanc
- Institute of Advanced Technologies in Living Sciences, CNRS - USR3505, Toulouse, France.,University of Toulouse, Institute of Research in Informatics of Toulouse, CNRS - UMR5505, Toulouse, France
| |
Collapse
|
32
|
161533 TriKE stimulates NK-cell function to overcome myeloid-derived suppressor cells in MDS. Blood Adv 2019; 2:1459-1469. [PMID: 29941459 DOI: 10.1182/bloodadvances.2017012369] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 05/18/2018] [Indexed: 12/24/2022] Open
Abstract
Myelodysplastic syndrome (MDS) is a clonal heterogeneous stem cell disorder driven by multiple genetic and epigenetic alterations resulting in ineffective hematopoiesis. MDS has a high frequency of immune suppressors, including myeloid-derived suppressor cells (MDSCs), that collectively result in a poor immune response. MDSCs in MDS patients express CD155 that ligates the T-cell immunoreceptor with immunoglobulin and ITIM domain (TIGIT) and delivers an inhibitory signal to natural killer (NK) cells. To mediate a productive immune response against MDS, negative regulatory checkpoints, like TIGIT, expressed on MDS NK cells must be overcome. NK cells can be directed to lyse MDS cells by bispecific killer engagers (BiKEs) that ligate CD16 on NK cells and CD33 on MDS cells. However, such CD16 × CD33 (1633) BiKEs do not induce the proliferative response in MDS NK cells needed to sustain their function. Here, we show that the addition of an NK stimulatory cytokine, interleukin-15 (IL-15), into the BiKE platform leads to productive IL-15 signaling without TIGIT upregulation on NK cells from MDS patients. Lower TIGIT expression allowed NK cells to resist MDSC inhibition. When compared with 1633 BiKE, 161533 trispecific killer engager (TriKE)-treated NK cells demonstrated superior killing kinetics associated with increased STAT5 phosphorylation. Furthermore, 161533 TriKE-treated MDS NK cells had higher proliferation and enhanced NK-cell function than 1633 BiKE-treated cells without the IL-15 linker. Collectively, our data demonstrate novel characteristics of the 161533 TriKE that support its application as an immunotherapeutic agent for MDS patients.
Collapse
|
33
|
Le Saux G, Schvartzman M. Advanced Materials and Devices for the Regulation and Study of NK Cells. Int J Mol Sci 2019; 20:E646. [PMID: 30717370 PMCID: PMC6386824 DOI: 10.3390/ijms20030646] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/24/2019] [Accepted: 01/29/2019] [Indexed: 02/07/2023] Open
Abstract
Natural Killer (NK) cells are innate lymphocytes that contribute to immune protection by cytosis, cytokine secretion, and regulation of adaptive responses of T cells. NK cells distinguish between healthy and ill cells, and generate a cytotoxic response, being cumulatively regulated by environmental signals delivered through their diverse receptors. Recent advances in biomaterials and device engineering paved the way to numerous artificial microenvironments for cells, which produce synthetic signals identical or similar to those provided by the physiological environment. In this paper, we review recent advances in materials and devices for artificial signaling, which have been applied to regulate NK cells, and systematically study the role of these signals in NK cell function.
Collapse
Affiliation(s)
- Guillaume Le Saux
- Department of Materials Engineering, Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel.
| | - Mark Schvartzman
- Department of Materials Engineering, Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel.
| |
Collapse
|
34
|
Liu Y, Yang Q, Cao L, Xu F. Analysis of Leukocyte Behaviors on Microfluidic Chips. Adv Healthc Mater 2019; 8:e1801406. [PMID: 30672149 DOI: 10.1002/adhm.201801406] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 01/05/2019] [Indexed: 01/17/2023]
Abstract
The orchestration of massive leukocytes in the immune system protects humans from invading pathogens and abnormal cells in the body. So far, researches focusing on leukocyte behaviors are performed based on both in vivo and in vitro models. The in vivo animal models are usually less controllable due to their extreme complexity and nonignorable species issue. Therefore, many researchers turn to in vitro models. With the advances in micro/nanofabrication, the microfluidic chip has emerged as a novel platform for model construction in multiple biomedical research fields. Specifically, the microfluidic chip is used to study leukocyte behaviors, due to its incomparable advantages in high throughput, precise control, and flexible integration. Moreover, the small size of the microstructures on the microfluidic chip can better mimic the native microenvironment of leukocytes, which contributes to a more reliable recapitulation. Herein are reviewed the recent advances in microfluidic chip-based leukocyte behavior analysis to provide an overview of this field. Detailed discussions are specifically focused on host defense against pathogens, immunodiagnosis, and immunotherapy studies on microfluidic chips. Finally, the current technical challenges are discussed, as well as possible innovations in this field to improve the related applications.
Collapse
Affiliation(s)
- Yan Liu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education; School of Life Science and Technology; Xi'an Jiaotong University; Xi'an Shaanxi 710049 China
- Bioinspired Engineering and Biomechanics Center (BEBC); Xi'an Jiaotong University; Xi'an Shaanxi 710049 China
| | - Qingzhen Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education; School of Life Science and Technology; Xi'an Jiaotong University; Xi'an Shaanxi 710049 China
- Bioinspired Engineering and Biomechanics Center (BEBC); Xi'an Jiaotong University; Xi'an Shaanxi 710049 China
| | - Lei Cao
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education; School of Life Science and Technology; Xi'an Jiaotong University; Xi'an Shaanxi 710049 China
- Bioinspired Engineering and Biomechanics Center (BEBC); Xi'an Jiaotong University; Xi'an Shaanxi 710049 China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education; School of Life Science and Technology; Xi'an Jiaotong University; Xi'an Shaanxi 710049 China
- Bioinspired Engineering and Biomechanics Center (BEBC); Xi'an Jiaotong University; Xi'an Shaanxi 710049 China
| |
Collapse
|
35
|
Sinha N, Subedi N, Tel J. Integrating Immunology and Microfluidics for Single Immune Cell Analysis. Front Immunol 2018; 9:2373. [PMID: 30459757 PMCID: PMC6232771 DOI: 10.3389/fimmu.2018.02373] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 09/24/2018] [Indexed: 12/16/2022] Open
Abstract
The field of immunoengineering aims to develop novel therapies and modern vaccines to manipulate and modulate the immune system and applies innovative technologies toward improved understanding of the immune system in health and disease. Microfluidics has proven to be an excellent technology for analytics in biology and chemistry. From simple microsystem chips to complex microfluidic designs, these platforms have witnessed an immense growth over the last decades with frequent emergence of new designs. Microfluidics provides a highly robust and precise tool which led to its widespread application in single-cell analysis of immune cells. Single-cell analysis allows scientists to account for the heterogeneous behavior of immune cells which often gets overshadowed when conventional bulk study methods are used. Application of single-cell analysis using microfluidics has facilitated the identification of several novel functional immune cell subsets, quantification of signaling molecules, and understanding of cellular communication and signaling pathways. Single-cell analysis research in combination with microfluidics has paved the way for the development of novel therapies, point-of-care diagnostics, and even more complex microfluidic platforms that aid in creating in vitro cellular microenvironments for applications in drug and toxicity screening. In this review, we provide a comprehensive overview on the integration of microsystems and microfluidics with immunology and focus on different designs developed to decode single immune cell behavior and cellular communication. We have categorized the microfluidic designs in three specific categories: microfluidic chips with cell traps, valve-based microfluidics, and droplet microfluidics that have facilitated the ongoing research in the field of immunology at single-cell level.
Collapse
Affiliation(s)
- Nidhi Sinha
- Laboratory of Immunoengineering, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Nikita Subedi
- Laboratory of Immunoengineering, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Jurjen Tel
- Laboratory of Immunoengineering, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| |
Collapse
|
36
|
Walwyn-Brown K, Guldevall K, Saeed M, Pende D, Önfelt B, MacDonald AS, Davis DM. Human NK Cells Lyse Th2-Polarizing Dendritic Cells via NKp30 and DNAM-1. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 201:2028-2041. [PMID: 30120122 PMCID: PMC6139540 DOI: 10.4049/jimmunol.1800475] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 07/23/2018] [Indexed: 01/08/2023]
Abstract
Cross-talk between NK cells and dendritic cells (DCs) is important in Th1 immune responses, including antitumor immunity and responses to infections. DCs also play a crucial role in polarizing Th2 immunity, but the impact of NK cell-DC interactions in this context remains unknown. In this study, we stimulated human monocyte-derived DCs in vitro with different pathogen-associated molecules: LPS or polyinosinic-polycytidylic acid, which polarize a Th1 response, or soluble egg Ag from the helminth worm Schistosoma mansoni, a potent Th2-inducing Ag. Th2-polarizing DCs were functionally distinguishable from Th1-polarizing DCs, and both showed distinct morphology and dynamics from immature DCs. We then assessed the outcome of autologous NK cells interacting with these differently stimulated DCs. Confocal microscopy showed polarization of the NK cell microtubule organizing center and accumulation of LFA-1 at contacts between NK cells and immature or Th2-polarizing DCs but not Th1-polarizing DCs, indicative of the assembly of an activating immune synapse. Autologous NK cells lysed immature DCs but not DCs treated with LPS or polyinosinic-polycytidylic acid as reported previously. In this study, we demonstrated that NK cells also degranulated in the presence of Th2-polarizing DCs. Moreover, time-lapse live-cell microscopy showed that DCs that had internalized fluorescently labeled soluble egg Ag were efficiently lysed. Ab blockade of NK cell-activating receptors NKp30 or DNAM-1 abrogated NK cell lysis of Th2-polarizing DCs. Thus, these data indicate a previously unrecognized role of NK cell cytotoxicity and NK cell-activating receptors NKp30 and DNAM-1 in restricting the pool of DCs involved in Th2 immune responses.
Collapse
Affiliation(s)
- Katherine Walwyn-Brown
- Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9NT, United Kingdom
| | - Karolin Guldevall
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Solna, SE-106 91 Stockholm, Sweden
| | - Mezida Saeed
- Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9NT, United Kingdom
| | - Daniela Pende
- Laboratorio Immunologia, Istituto di Ricovero e Cura a Carattere Scientifico, Ospedale Policlinico San Martino, 16132 Genova, Italy; and
| | - Björn Önfelt
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Solna, SE-106 91 Stockholm, Sweden
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Andrew S MacDonald
- Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9NT, United Kingdom
| | - Daniel M Davis
- Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9NT, United Kingdom;
| |
Collapse
|
37
|
Irimia D, Wang X. Inflammation-on-a-Chip: Probing the Immune System Ex Vivo. Trends Biotechnol 2018; 36:923-937. [PMID: 29728272 PMCID: PMC6098972 DOI: 10.1016/j.tibtech.2018.03.011] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 03/23/2018] [Accepted: 03/26/2018] [Indexed: 01/02/2023]
Abstract
Inflammation is the typical result of activating the host immune system against pathogens, and it helps to clear microbes from tissues. However, inflammation can occur in the absence of pathogens, contributing to tissue damage and leading to disease. Understanding how immune cells coordinate their activities to initiate, modulate, and terminate inflammation is key to developing effective interventions to preserve health and combat diseases. Towards this goal, inflammation-on-a-chip tools provide unique features that greatly benefit the study of inflammation. They reconstitute tissue environments in microfabricated devices and enable real-time, high-resolution observations and quantification of cellular activities relevant to inflammation. We review here recent advances in inflammation-on-a-chip technologies and highlight the biological insights and clinical applications enabled by these emerging tools.
Collapse
Affiliation(s)
- Daniel Irimia
- BioMEMS Resource Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Shriners Burns Hospital, Boston, MA
| | - Xiao Wang
- BioMEMS Resource Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Shriners Burns Hospital, Boston, MA
| |
Collapse
|
38
|
Mishra HK, Pore N, Michelotti EF, Walcheck B. Anti-ADAM17 monoclonal antibody MEDI3622 increases IFNγ production by human NK cells in the presence of antibody-bound tumor cells. Cancer Immunol Immunother 2018; 67:1407-1416. [PMID: 29978334 PMCID: PMC6126979 DOI: 10.1007/s00262-018-2193-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 06/29/2018] [Indexed: 01/11/2023]
Abstract
Several clinically successful tumor-targeting mAbs induce NK cell effector functions. Human NK cells exclusively recognize tumor-bound IgG by the FcR CD16A (FcγRIIIA). Unlike other NK cell activating receptors, the cell surface density of CD16A can be rapidly downregulated in a cis manner by the metalloproteinase ADAM17 following NK cell stimulation in various manners. CD16A downregulation takes place in cancer patients and this may affect the efficacy of tumor-targeting mAbs. We examined the effects of MEDI3622, a human mAb and potent ADAM17 inhibitor, on NK cell activation by antibody-bound tumor cells. MEDI3622 effectively blocked ADAM17 function in NK cells and caused a marked increase in their production of IFNγ. This was observed for NK cells exposed to different tumor cell lines and therapeutic antibodies, and over a range of effector/target ratios. The augmented release of IFNγ by NK cells was reversed by a function-blocking CD16A mAb. In addition, NK92 cells, a human NK cell line that lacks endogenous FcγRs, expressing a recombinant non-cleavable version of CD16A released significantly higher levels of IFNγ than NK92 cells expressing equivalent levels of wildtype CD16A. Taken together, our data show that MEDI3622 enhances the release of IFNγ by NK cells engaging antibody-bound tumor cells by blocking the shedding of CD16A. These findings support ADAM17 as a dynamic inhibitory checkpoint of the potent activating receptor CD16A, which can be targeted by MEDI3622 to potentially increase the efficacy of anti-tumor therapeutic antibodies.
Collapse
Affiliation(s)
- Hemant K Mishra
- Department of Veterinary and Biomedical Sciences, University of Minnesota, 295B AS/VM Bldg., 1988 Fitch Avenue, St. Paul, MN, 55108, USA
| | - Nabendu Pore
- Oncology Research, MedImmune, LLC, Gaithersburg, USA
| | - Emil F Michelotti
- Oncology Research, MedImmune, LLC, Gaithersburg, USA
- NIC, NIH, Bethesda, MD, 20892, USA
| | - Bruce Walcheck
- Department of Veterinary and Biomedical Sciences, University of Minnesota, 295B AS/VM Bldg., 1988 Fitch Avenue, St. Paul, MN, 55108, USA.
| |
Collapse
|
39
|
Park H, Kim H, Doh J. Multifunctional Microwell Arrays for Single Cell Level Functional Analysis of Lymphocytes. Bioconjug Chem 2017; 29:672-679. [DOI: 10.1021/acs.bioconjchem.7b00620] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
40
|
Ke LY, Kuo ZK, Chen YS, Yeh TY, Dong M, Tseng HW, Liu CH. Cancer immunotherapy μ-environment LabChip: taking advantage of optoelectronic tweezers. LAB ON A CHIP 2017; 18:106-114. [PMID: 29211085 DOI: 10.1039/c7lc00963a] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
A cancer immunotherapy μ-environment LabChip, equipped with titanium oxide phthalocyanine (TiOPc)-based optoelectronic tweezers (OET) to achieve direct cell-cell contact, can be used to study the interaction between immune cells and other cells for real-time analysis of NK cells' behavior. In microfluidic devices, it is difficult to solve dead zone problems and observe dynamic cell-cell interactions. We have created a stable and static culture μ-environment which can enhance NK cell activities. In addition, OET is used to solve dead zone problems by manipulating a single cell into four-leaf-clover-shaped (FLCS) microwells made of poly(ethylene glycol) diacrylate (PEG-DA) through optofluidic maskless lithography, causing direct cell-cell contact. Our design reconstructed an in vitro human immune system for the study of dynamic immunological response. When the NK cells came into contact with the target cells in the μ-environment LabChip, we observed that the target cells showed apoptotic characteristics (i.e. cell shrinkage and blebbing within 2 h and then die within 3 h). In addition, our μ-environment LabChip demonstrated higher NK cell activity compared with conventional analysis. We have created an innovative cancer immunotherapy μ-environment LabChip to provide a stable and static μ-environment for cell-cell interaction study. Furthermore, our μ-environment LabChip showed the potential to enhance NK cell activity and to study immunological interactions between immune cells and cancer cells dynamically.
Collapse
Affiliation(s)
- Ling-Yi Ke
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu City 30013, Taiwan, R.O.C.
| | | | | | | | | | | | | |
Collapse
|
41
|
Xu Y, Zhou S, Lam YW, Pang SW. Dynamics of Natural Killer Cells Cytotoxicity in Microwell Arrays with Connecting Channels. Front Immunol 2017; 8:998. [PMID: 28861090 PMCID: PMC5562680 DOI: 10.3389/fimmu.2017.00998] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Accepted: 08/04/2017] [Indexed: 01/22/2023] Open
Abstract
Natural killer (NK) cells serve an important role in immune system by recognizing and killing the potentially malignant cells without antigen sensitization, and could be promising in cancer therapy. We have designed and fabricated microwell arrays with microchannel connections in polydimethylsiloxane (PDMS) substrates to study the interaction dynamics of NK-92MI cells with MCF7 breast cancer cells using time-lapse imaging by fluorescence microscopy for 15 h. Although cell seeding density was the same, NK cell cytotoxicity was found to be higher in larger microwells, which is manifested as increased target death ratio from 13.7 ± 3.1 to 46.3 ± 3.3% and shorter triggering time of first target lysis from 502 ± 49 to 391 ± 63 min in 150 μm × 150 μm microwells comparing to 50 μm × 50 μm wells in 15 h. Mirochannel connection between adjacent microwells of the same size increased the overall target death ratio by >10%, while connection between microwells of different sizes led to significantly increased target death ratio and delayed first target lysis in smaller microwells. Our findings reveal unique cell interaction dynamics, such as initiation and stimulation, of NK cell cytotoxicity in a confined microenvironment, which is different from population-based study, and the results could lead to a better understanding of the dynamics of NK cell cytotoxicity.
Collapse
Affiliation(s)
- Yuanhao Xu
- Department of Electronic Engineering, City University of Hong Kong, Kowloon, Hong Kong.,Center for Biosystems, Neuroscience, and Nanotechnology, City University of Hong Kong, Kowloon, Hong Kong
| | - Shufan Zhou
- Department of Electronic Engineering, City University of Hong Kong, Kowloon, Hong Kong.,Center for Biosystems, Neuroscience, and Nanotechnology, City University of Hong Kong, Kowloon, Hong Kong
| | - Yun Wah Lam
- Center for Biosystems, Neuroscience, and Nanotechnology, City University of Hong Kong, Kowloon, Hong Kong.,Department of Biology and Chemistry, City University of Hong Kong, Kowloon, Hong Kong
| | - Stella W Pang
- Department of Electronic Engineering, City University of Hong Kong, Kowloon, Hong Kong.,Center for Biosystems, Neuroscience, and Nanotechnology, City University of Hong Kong, Kowloon, Hong Kong
| |
Collapse
|
42
|
Li Y, Jang JH, Wang C, He B, Zhang K, Zhang P, Vu T, Qin L. Microfluidics Cell Loading-Dock System: Ordered Cellular Array for Dynamic Lymphocyte-Communication Study. ACTA ACUST UNITED AC 2017; 1:e1700085. [PMID: 32646193 DOI: 10.1002/adbi.201700085] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 07/12/2017] [Indexed: 12/26/2022]
Abstract
It remains a great challenge to establish a high-throughput platform that can explore the interactions among multiple lymphocytes (>2 cells) and retrieve the interested cells for downstream analysis. This study demonstrates a microfluidics cell loading-dock system (Cell-Dock) to enclose multiple cells in 1D, 2D, and 3D chambers with high throughput and efficiency and single-cell accuracy. The loading efficiencies of 95%, 85%, and 74% for one-, three-, and five-cell systems are achieved, respectively. The Cell-Dock system provides precise and dynamic cell packing models to facilitate lymphocyte-interaction studies. The results demonstrate that individual natural killer (NK) cells may function independently rather than cooperate to lyse target cells in the defined microenvironment. Furthermore, the strong/weak NK cells are retrieved based on their on-chip cytotoxicity and mRNA sequencing is conducted to find the possible mechanisms for "serial killing," an important but unsolved issue. This study finds that the stronger NK cells overexpress multiple genes involved in cytotoxicity and adhesion molecules (including the well-known ICAM1 and seldom reported B4GALT1) might play important roles in the regulation of NK cytolysis.
Collapse
Affiliation(s)
- Ying Li
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA.,Department of Cell and Developmental Biology, Weill Medical College of Cornell University, New York, NY, 10065, USA
| | - Joon Hee Jang
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA.,Department of Cell and Developmental Biology, Weill Medical College of Cornell University, New York, NY, 10065, USA
| | - Crystal Wang
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA.,Department of Cell and Developmental Biology, Weill Medical College of Cornell University, New York, NY, 10065, USA
| | - Bangshun He
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA.,Department of Cell and Developmental Biology, Weill Medical College of Cornell University, New York, NY, 10065, USA.,Central Laboratory, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Kai Zhang
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA.,Department of Cell and Developmental Biology, Weill Medical College of Cornell University, New York, NY, 10065, USA
| | - Pengchao Zhang
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA.,Department of Cell and Developmental Biology, Weill Medical College of Cornell University, New York, NY, 10065, USA
| | - Timothy Vu
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA.,Department of Cell and Developmental Biology, Weill Medical College of Cornell University, New York, NY, 10065, USA.,Department of Bioengineering, Rice University, Houston, TX, 77005, USA
| | - Lidong Qin
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA.,Department of Cell and Developmental Biology, Weill Medical College of Cornell University, New York, NY, 10065, USA
| |
Collapse
|
43
|
Mukherjee M, Mace EM, Carisey AF, Ahmed N, Orange JS. Quantitative Imaging Approaches to Study the CAR Immunological Synapse. Mol Ther 2017; 25:1757-1768. [PMID: 28663103 PMCID: PMC5542801 DOI: 10.1016/j.ymthe.2017.06.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 06/03/2017] [Accepted: 06/04/2017] [Indexed: 01/11/2023] Open
Abstract
The lytic immunological synapse (IS) is a discrete structural entity formed after the ligation of specific activating receptors that leads to the destruction of a cancerous cell. The formation of an effector cell IS in cytotoxic T lymphocytes or natural killer cells is a hierarchical and stepwise rearrangement of structural and signaling components and targeted release of the contents of lytic granules. While recent advances in the generation and testing of cytotoxic lymphocytes expressing chimeric antigen receptors (CARs) has demonstrated their efficacy in the targeted lysis of tumor targets, the contribution and dynamics of IS components have not yet been extensively investigated in the context of engineered CAR cells. Understanding the biology of the CAR IS will be a powerful approach to efficiently guide the engineering of new CARs and help identify mechanistic problems in existing CARs. Here, we review the formation of the lytic IS and describe quantitative imaging-based measurements using multiple microscopy techniques at a single cell level that can be used in conjunction with established population-based assays to provide insight into the important cytotoxic function of CAR cells. The inclusion of this approach in the pipeline of CAR product design could be a novel and valuable innovation for the field.
Collapse
MESH Headings
- Animals
- Antigens/chemistry
- Antigens/immunology
- Antigens/metabolism
- Biotechnology
- Cytotoxicity, Immunologic
- Humans
- Immunological Synapses/immunology
- Immunological Synapses/metabolism
- Immunotherapy/methods
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Microscopy/methods
- Molecular Imaging/methods
- Receptors, Antigen, T-Cell/chemistry
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- Recombinant Fusion Proteins
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
Collapse
Affiliation(s)
- Malini Mukherjee
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77025, USA; Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77025, USA; Center for Human Immunobiology, Texas Children's Hospital, Houston, TX 77030, USA.
| | - Emily M Mace
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77025, USA; Center for Human Immunobiology, Texas Children's Hospital, Houston, TX 77030, USA
| | - Alexandre F Carisey
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77025, USA; Center for Human Immunobiology, Texas Children's Hospital, Houston, TX 77030, USA
| | - Nabil Ahmed
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77025, USA; Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77025, USA
| | - Jordan S Orange
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77025, USA; Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77025, USA; Center for Human Immunobiology, Texas Children's Hospital, Houston, TX 77030, USA
| |
Collapse
|
44
|
Kim JS, Shin BR, Lee HK, Lee JH, Kim KH, Choi JE, Ji AY, Hong JT, Kim Y, Han SB. Cd226-/- natural killer cells fail to establish stable contacts with cancer cells and show impaired control of tumor metastasis in vivo. Oncoimmunology 2017; 6:e1338994. [PMID: 28920003 DOI: 10.1080/2162402x.2017.1338994] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 05/30/2017] [Accepted: 06/02/2017] [Indexed: 12/21/2022] Open
Abstract
CD226 is an activating receptor expressed on natural killer (NK) cells, CD8+ T cells, and other immune cells. Upon binding to its ligands expressed on target cells, CD226 activates intracellular signaling that triggers cytokine production and degranulation in NK cells. However, the role of CD226 in contact dynamics between NK and cancer cells has remained unclear. Our time-lapse images showed that individual wild-type CD226+ NK cells contacted B16F10 melanoma cells for 23.7 min, but Cd226-/- NK cells only for 12.8 min, although both NK cell subsets showed equal contact frequency over 4 h. On the surface of B16F10 cells, CD226+ cells stayed at the same site with oscillating movement (named stable contact), while Cd226-/- NK cells moved around at a velocity of 4 μm/min (named unstable contact). Consequently, Cd226-/- NK cells did not kill B16F10 cells in vitro and did not inhibit their metastasis into the lung in vivo. Taken together, our data demonstrate that CD226 enables prolonged stable interaction between NK and cancer cells, which is needed for efficient killing of cancer cells.
Collapse
Affiliation(s)
- Ji Sung Kim
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Bo Ram Shin
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Hong Kyung Lee
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Jae Hee Lee
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Ki Hun Kim
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Jeong Eun Choi
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - A Young Ji
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Youngsoo Kim
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Sang-Bae Han
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| |
Collapse
|