1
|
Kumar ARK, Low J, Lim J, Myint B, Sun X, Wu L, Cheng HS, Yip S, Ming Cheng CZ, Manoharan T, Quek YJ, Shou Y, Tian JS, Ng YY, Gascoigne NRJ, Tan NS, Sugimura R, Chia G, Sze Cheung AM, Yawata M, Tay A. Non-viral, high throughput genetic engineering of primary immune cells using nanostraw-mediated transfection. Biomaterials 2025; 317:123079. [PMID: 39842078 DOI: 10.1016/j.biomaterials.2024.123079] [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: 11/14/2024] [Revised: 12/30/2024] [Accepted: 12/30/2024] [Indexed: 01/24/2025]
Abstract
Transfection of proteins, mRNA, and chimeric antigen receptor (CAR) transgenes into immune cells remains a critical bottleneck in cell manufacturing. Current methods, such as viruses and bulk electroporation, are hampered by low transfection efficiency, unintended transgene integration, and significant cell perturbation. The Nanostraw Electro-actuated Transfection (NExT) technology offers a solution by using high aspect-ratio nanostraws and localized electric fields to precisely deliver biomolecules into cells with minimal disruption. We demonstrate that NExT can deliver proteins, polysaccharides, and mRNA into primary human CD8+ and CD4+ T cells, and achieve CRISPR/Cas9 gene knockout of CXCR4 and TRAC in CD8+ T cells. We showcase NExT's versatility across a range of primary human immune cells, including CD4+ T cells, γδ-T cells, dendritic cells, NK cells, Treg cells, macrophages, and neutrophils. Finally, we developed a scalable, high-throughput multiwell NExT system capable of transfecting over 14 million cells and delivering diverse cargoes into multiple cell types from various donors simultaneously. This technology holds promise for streamlining high-throughput screening of allogeneic donors and reducing optimization costs for large-scale CAR-immune cell transfection.
Collapse
Affiliation(s)
- Arun R K Kumar
- Institute of Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore; Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Jessalyn Low
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Jet Lim
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Ba Myint
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Xinhong Sun
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Ling Wu
- Immunology Translational Research Programme and Department of Microbiology and Immunology, National University of Singapore, Singapore, 117545, Singapore
| | - Hong Sheng Cheng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore; School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Sophronia Yip
- Li Ka Shing Faculty of Medicine, School of Biomedical Sciences, University of Hong Kong, Hong Kong SAR, China; Centre for Translational Stem Cell Biology, Hong Kong SAR, China
| | - Cyrus Zai Ming Cheng
- Institute of Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore; Department of Pharmacy, National University of Singapore, Singapore, 117559, Singapore
| | - Thamizhanban Manoharan
- Institute of Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore; Department of Pharmacy, National University of Singapore, Singapore, 117559, Singapore
| | - Ying Jie Quek
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Yufeng Shou
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Johann Shane Tian
- Institute of Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore; Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Yu Yang Ng
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Nicholas R J Gascoigne
- Immunology Translational Research Programme and Department of Microbiology and Immunology, National University of Singapore, Singapore, 117545, Singapore
| | - Nguan Soon Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore; School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Rio Sugimura
- Li Ka Shing Faculty of Medicine, School of Biomedical Sciences, University of Hong Kong, Hong Kong SAR, China; Centre for Translational Stem Cell Biology, Hong Kong SAR, China
| | - Gloryn Chia
- Institute of Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore; Department of Pharmacy, National University of Singapore, Singapore, 117559, Singapore
| | - Alice Man Sze Cheung
- Department of Haematology, Singapore General Hospital, Singapore, 169608, Singapore; SingHealth Duke-NUS Medicine Academic Clinical Program, Duke-NUS Medical School, Singapore, 168753, Singapore
| | - Makoto Yawata
- Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, 119077, Singapore
| | - Andy Tay
- Institute of Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore; Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore; NUS Tissue Engineering Program, National University of Singapore, Singapore, 117510, Singapore.
| |
Collapse
|
2
|
Sun N, Wang C, Edwards W, Wang Y, Lu XL, Gu C, McLennan S, Shangaris P, Qi P, Mastronicola D, Scottà C, Lombardi G, Chiappini C. Nanoneedle-Based Electroporation for Efficient Manufacturing of Human Primary Chimeric Antigen Receptor Regulatory T-Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2416066. [PMID: 40231643 DOI: 10.1002/advs.202416066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 04/03/2025] [Indexed: 04/16/2025]
Abstract
Regulatory T cells (Tregs) play a crucial role in moderating immune responses offering promising therapeutic options for autoimmune diseases and allograft rejection. Genetically engineering Tregs with chimeric antigen receptors (CARs) enhances their targeting specificity and efficacy. With non-viral transfection methods suffering from low efficiency and reduced cell viability, viral transduction is currently the only viable approach for GMP-compliant CAR-Treg production. However, viral transduction raises concerns over immunogenicity, insertional mutagenesis risk, and high costs, which limit clinical scalability. This study introduces a scalable nanoneedle electroporation (nN-EP) platform for GMP-compatible transfection of HLA-A2-specific CAR plasmids into primary human Tregs. The nN-EP system achieves 43% transfection efficiency, outperforming viral transduction at multiplicity of infection 1 by twofold. Importantly, nN-EP preserves Treg viability, phenotype and proliferative capacity. HLA-A2-specific CAR-Tregs generated using nN-EP show specific activation and superior suppressive function compared to polyclonal or virally transduced Tregs in the presence of HLA-A2 expressing antigen presenting cells. These findings underscore the potential of nN-EP as a GMP-suitable method for CAR-Treg production, enabling broader clinical application in immune therapies.
Collapse
Affiliation(s)
- Ningjia Sun
- Centre for Craniofacial and Regenerative Biology, King's College London, London, SE1 9RT, UK
| | - Cong Wang
- Centre for Craniofacial and Regenerative Biology, King's College London, London, SE1 9RT, UK
- London Centre for Nanotechnology, King's College London, London, WC2R 2LS, UK
- Wenzhou Eye Valley Innovation Center, Eye Hospital, Wenzhou Medical University, Zhejiang, 325035, China
| | - William Edwards
- Centre for Craniofacial and Regenerative Biology, King's College London, London, SE1 9RT, UK
| | - Yikai Wang
- Centre for Craniofacial and Regenerative Biology, King's College London, London, SE1 9RT, UK
- London Centre for Nanotechnology, King's College London, London, WC2R 2LS, UK
| | - Xiangrong L Lu
- Centre for Craniofacial and Regenerative Biology, King's College London, London, SE1 9RT, UK
- Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK
| | - Chenlei Gu
- Centre for Craniofacial and Regenerative Biology, King's College London, London, SE1 9RT, UK
- London Centre for Nanotechnology, King's College London, London, WC2R 2LS, UK
| | - Samuel McLennan
- Centre for Craniofacial and Regenerative Biology, King's College London, London, SE1 9RT, UK
- London Centre for Nanotechnology, King's College London, London, WC2R 2LS, UK
| | - Panicos Shangaris
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, London, SE1 7EH, UK
- School of Life Course & Population Sciences, 10th Floor North Wing, St Thomas' Hospital, King's College London, London, SE1 7EH, UK
- Harris Birthright Research Centre for Fetal Medicine, King's College London, London, SE1 7EH, UK
| | - Peng Qi
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, London, SE1 7EH, UK
| | - Daniela Mastronicola
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, London, SE1 7EH, UK
| | - Cristiano Scottà
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, London, SE1 7EH, UK
- Department of Biosciences, Centre for Inflammation Research and Translational Medicine, Brunel University London, London, UB8 3PH, UK
| | - Giovanna Lombardi
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, London, SE1 7EH, UK
| | - Ciro Chiappini
- Centre for Craniofacial and Regenerative Biology, King's College London, London, SE1 9RT, UK
- London Centre for Nanotechnology, King's College London, London, WC2R 2LS, UK
| |
Collapse
|
3
|
Li Z, Sarikhani E, Prayotamornkul S, Meganathan DP, Jahed Z, Shi L. Multimodal Imaging Unveils the Impact of Nanotopography on Cellular Metabolic Activities. CHEMICAL & BIOMEDICAL IMAGING 2024; 2:825-834. [PMID: 39735831 PMCID: PMC11672213 DOI: 10.1021/cbmi.4c00051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 12/31/2024]
Abstract
Nanoscale surface topography is an effective approach in modulating cell-material interactions, significantly impacting cellular and nuclear morphologies, as well as their functionality. However, the adaptive changes in cellular metabolism induced by the mechanical and geometrical microenvironment of the nanotopography remain poorly understood. In this study, we investigated the metabolic activities in cells cultured on engineered nanopillar substrates by using a label-free multimodal optical imaging platform. This multimodal imaging platform, integrating two photon fluorescence (TPF) and stimulated Raman scattering (SRS) microscopy, allowed us to directly visualize and quantify metabolic activities of cells in 3D at the subcellular scale. We discovered that the nanopillar structure significantly reduced the cell spreading area and circularity compared to flat surfaces. Nanopillar-induced mechanical cues significantly modulate cellular metabolic activities with variations in nanopillar geometry further influencing these metabolic processes. Cells cultured on nanopillars exhibited reduced oxidative stress, decreased protein and lipid synthesis, and lower lipid unsaturation in comparison to those on flat substrates. Hierarchical clustering also revealed that pitch differences in the nanopillar had a more significant impact on cell metabolic activity than diameter variations. These insights improve our understanding of how engineered nanotopographies can be used to control cellular metabolism, offering possibilities for designing advanced cell culture platforms which can modulate cell behaviors and mimic natural cellular environment and optimize cell-based applications. By leveraging the unique metabolic effects of nanopillar arrays, one can develop more effective strategies for directing the fate of cells, enhancing the performance of cell-based therapies, and creating regenerative medicine applications.
Collapse
Affiliation(s)
- Zhi Li
- Shu
Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, California 92093, United States
| | - Einollah Sarikhani
- Aiiso
Yufeng Li Family Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, California 92093, United States
| | - Sirasit Prayotamornkul
- Shu
Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, California 92093, United States
| | - Dhivya Pushpa Meganathan
- Shu
Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, California 92093, United States
| | - Zeinab Jahed
- Shu
Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, California 92093, United States
- Aiiso
Yufeng Li Family Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, California 92093, United States
| | - Lingyan Shi
- Shu
Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, California 92093, United States
- Aiiso
Yufeng Li Family Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, California 92093, United States
- Electrical
and Computer Engineering, University of
California San Diego, La Jolla, California 92093, United States
- Institute
of Engineering in Medicine, University of
California San Diego, La Jolla, California 92093, United States
- Synthetic
Biology Institute, University of California
San Diego, La Jolla, California 92093, United States
| |
Collapse
|
4
|
Mahalanabish A, Huang SH, Tulegenov D, Shvets G. Infrared Spectroscopy of Live Cells Using High-Aspect-Ratio Metal-on-Dielectric Metasurfaces. NANO LETTERS 2024; 24:11607-11614. [PMID: 39248258 DOI: 10.1021/acs.nanolett.4c03155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/10/2024]
Abstract
Fourier transform infrared (FTIR) spectroscopy is widely used for molecular analysis. However, for the materials situated in an aqueous environment, a precondition for live biological objects such as cells, transmission-based FTIR is prevented by strong water absorption of mid-infrared (MIR) light. Reflection-based cellular assays using internal reflection elements (IREs) such as high-index prisms or flat plasmonic metasurfaces mitigate these issues but suffer from a shallow probing volume localized near the plasma membrane. Inspired by the recent introduction of high-aspect-ratio nanostructures as a novel platform for manipulating cellular behavior, we demonstrate that the integration of plasmonic metasurfaces with tall dielectric nanostructures dramatically enhances the sensing capabilities of FTIR spectroscopy. We also demonstrate the ability of a metal-on-dielectric metasurface to transduce intracellular processes, such as protein translocation to high-curvature membrane regions during cell adhesion, into interpretable spectral signatures of the reflected light.
Collapse
Affiliation(s)
- Aditya Mahalanabish
- School of Applied and Engineering Physics, Cornell University, Ithaca, New York 14853, United States
| | - Steven H Huang
- School of Applied and Engineering Physics, Cornell University, Ithaca, New York 14853, United States
| | - Dias Tulegenov
- School of Applied and Engineering Physics, Cornell University, Ithaca, New York 14853, United States
| | - Gennady Shvets
- School of Applied and Engineering Physics, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
5
|
Sarikhani E, Meganathan DP, Larsen AKK, Rahmani K, Tsai CT, Lu CH, Marquez-Serrano A, Sadr L, Li X, Dong M, Santoro F, Cui B, Klausen LH, Jahed Z. Engineering the Cellular Microenvironment: Integrating Three-Dimensional Nontopographical and Two-Dimensional Biochemical Cues for Precise Control of Cellular Behavior. ACS NANO 2024; 18:19064-19076. [PMID: 38978500 PMCID: PMC11271182 DOI: 10.1021/acsnano.4c03743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 07/10/2024]
Abstract
The development of biomaterials capable of regulating cellular processes and guiding cell fate decisions has broad implications in tissue engineering, regenerative medicine, and cell-based assays for drug development and disease modeling. Recent studies have shown that three-dimensional (3D) nanoscale physical cues such as nanotopography can modulate various cellular processes like adhesion and endocytosis by inducing nanoscale curvature on the plasma and nuclear membranes. Two-dimensional (2D) biochemical cues such as protein micropatterns can also regulate cell function and fate by controlling cellular geometries. Development of biomaterials with precise control over nanoscale physical and biochemical cues can significantly influence programming cell function and fate. In this study, we utilized a laser-assisted micropatterning technique to manipulate the 2D architectures of cells on 3D nanopillar platforms. We performed a comprehensive analysis of cellular and nuclear morphology and deformation on both nanopillar and flat substrates. Our findings demonstrate the precise engineering of single cell architectures through 2D micropatterning on nanopillar platforms. We show that the coupling between the nuclear and cell shape is disrupted on nanopillar surfaces compared to flat surfaces. Furthermore, our results suggest that cell elongation on nanopillars enhances nanopillar-induced endocytosis. We believe our platform serves as a versatile tool for further explorations into programming cell function and fate through combined physical cues that create nanoscale curvature on cell membranes and biochemical cues that control the geometry of the cell.
Collapse
Affiliation(s)
- Einollah Sarikhani
- Department
of NanoEngineering, University of California
San Diego, La Jolla ,California 92093, United States
| | - Dhivya Pushpa Meganathan
- Department
of NanoEngineering, University of California
San Diego, La Jolla ,California 92093, United States
| | | | - Keivan Rahmani
- Department
of NanoEngineering, University of California
San Diego, La Jolla ,California 92093, United States
| | - Ching-Ting Tsai
- Department
of Chemistry, Stanford University, Stanford ,California 94305, United States
| | - Chih-Hao Lu
- Department
of Chemistry, Stanford University, Stanford ,California 94305, United States
| | - Abel Marquez-Serrano
- Department
of NanoEngineering, University of California
San Diego, La Jolla ,California 92093, United States
| | - Leah Sadr
- Department
of NanoEngineering, University of California
San Diego, La Jolla ,California 92093, United States
| | - Xiao Li
- Department
of Chemistry, Stanford University, Stanford ,California 94305, United States
| | - Mingdong Dong
- Interdisciplinary
Nanoscience Center (iNANO), Aarhus University, Aarhus C 8000, Denmark
| | - Francesca Santoro
- Center
for Advanced Biomaterials for Healthcare, Tissue Electronics, Instituto Italiano di Tecnologia, Naples 80125, Italy
- Faculty
of Electrical Engineering and IT, RWTH, Aachen 52074, Germany
- Institute
for Biological Information Processing-Bioelectronics, Forschungszentrum
Juelich, Julich 52428, Germany
| | - Bianxiao Cui
- Department
of Chemistry, Stanford University, Stanford ,California 94305, United States
| | | | - Zeinab Jahed
- Department
of NanoEngineering, University of California
San Diego, La Jolla ,California 92093, United States
- Department
of Bioengineering, University of California
San Diego, La Jolla ,California 92093, United States
| |
Collapse
|
6
|
Yang D, Li C, Kong Y, Pei Y, Miao B, Dai G, Ding P, Shi P, Wang Z, Pei R. Deciphering the Temporal-Spatial Interactive Heterogeneity of Long Non-Coding RNAs and RNA-Binding Proteins in Living Cells at Single-Cell Resolution. J Am Chem Soc 2024. [PMID: 39016781 DOI: 10.1021/jacs.4c05205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
The investigation of long noncoding RNAs (lncRNAs) and RNA binding proteins (RBPs) interactions in living cell holds great significance for elucidating their critical roles in a variety of biological activities, but limited techniques are available to profile the temporal-spatial dynamic heterogeneity. Here, we introduced a molecular beacon-functionalized nanoneedle array designed for spatially resolved profiling of lncRNA-RBP interactions (Nano-SpatiaLR). A nanoneedle array modified with a molecular beacon is employed to selectively isolate specific intracellular lncRNAs and their associated RBPs without affecting cell viability. The RBPs are then in situ analyzed with a fluorescent labeled antibody and colocalized with lncRNA signals to get a quantitative measurement of their dynamic interactions. Additionally, leveraging the spatial distribution and nanoscale modality of the nanoneedle array, this technique provides the spatial heterogeneity information on cellular lncRNA-RBPs interaction at single cell resolution. In this study, we tracked the temporal-spatial interactive heterogeneity dynamics of lncRNA-RBPs interaction within living cells across different biological progresses. Our findings demonstrated that the interactions between lncRNA HOTAIR and RBPs EZH2 and LSD1 undergo significant changes in response to drug treatments, particularly in tumor cells. Moreover, these interactions become more intensified as tumor cells aggregate during the proliferation process.
Collapse
Affiliation(s)
- Deyuan Yang
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Cheng Li
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Yutong Kong
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yian Pei
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
- Duke Kunshan University Kunshan 215316, China
| | - Bing Miao
- Key Laboratory of Multifunctional Nanomaterials and Smart Systems, Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Gaole Dai
- Suzhou Institute of Systems Medicine, Suzhou 215123, China
| | - Pi Ding
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Peng Shi
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering, Hong Kong Science Park, Shatin, Hong Kong 999077, China
- Department of Biomedical Engineering, City University of Hong Kong Kowloon, Shatin, Hong Kong 999077, China
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen 518000, China
| | - Zixun Wang
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Renjun Pei
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| |
Collapse
|
7
|
Chen Y, Shokouhi AR, Voelcker NH, Elnathan R. Nanoinjection: A Platform for Innovation in Ex Vivo Cell Engineering. Acc Chem Res 2024; 57:1722-1735. [PMID: 38819691 PMCID: PMC11191407 DOI: 10.1021/acs.accounts.4c00190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 06/01/2024]
Abstract
In human cells, intracellular access and therapeutic cargo transport, including gene-editing tools (e.g., CRISPR-Cas9 and transposons), nucleic acids (e.g., DNA, mRNA, and siRNA), peptides, and proteins (e.g., enzymes and antibodies), are tightly constrained to ensure healthy cell function and behavior. This principle is exemplified in the delivery mechanisms of chimeric antigen receptor (CAR)-T cells for ex-vivo immunotherapy. In particular, the clinical success of CAR-T cells has established a new standard of care by curing previously incurable blood cancers. The approach involves the delivery, typically via the use of electroporation (EP) and lentivirus, of therapeutic CAR genes into a patient's own T cells, which are then engineered to express CARs that target and combat their blood cancer. But the key difficulty lies in genetically manipulating these cells without causing irreversible damage or loss of function─all the while minimizing complexities of manufacturing, safety concerns, and costs, and ensuring the efficacy of the final CAR-T cell product.Nanoinjection─the process of intracellular delivery using nanoneedles (NNs)─is an emerging physical delivery route that efficiently negotiates the plasma membrane of many cell types, including primary human T cells. It occurs with minimal perturbation, invasiveness, and toxicity, with high efficiency and throughput at high spatial and temporal resolutions. Nanoinjection promises greatly improved delivery of a broad range of therapeutic cargos with little or no damage to those cargos. A nanoinjection platform allows these cargos to function in the intracellular space as desired. The adaptability of nanoinjection platforms is now bringing major advantages in immunomodulation, mechanotransduction, sampling of cell states (nanobiopsy), controlled intracellular interrogation, and the primary focus of this account─intracellular delivery and its applications in ex vivo cell engineering. Mechanical nanoinjection typically exerts direct mechanical force on the cell membrane, offering a straightforward route to improve membrane perturbation by the NNs and subsequent transport of genetic cargo into targeted cell type (adherent or suspension cells). By contrast, electroactive nanoinjection is controlled by coupling NNs with an electric field─a new route for activating electroporation (EP) at the nanoscale─allowing a dramatic reduction of the applied voltage to a cell and so minimizing post-EP damage to cells and cargo, and overcoming many of the limitations of conventional bulk EP. Nanoinjection transcends mere technique; it is an approach to cell engineering ex vivo, offering the potential to endow cells with new, powerful features such as generating chimeric antigen receptor (CAR)-T cells for future CAR-T cell technologies. We first discuss the manufacturing of NN devices (Section 2), then delve into nanoinjection-mediated cell engineering (Section 3), nanoinjection mechanisms and interfacing methodologies (Section 4), and emerging applications in using nanoinjection to create functional CAR-T cells (Section 5).
Collapse
Affiliation(s)
- Yaping Chen
- Oujiang
Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain
Health), Institute of Aging, Key Laboratory of Alzheimer’s
Disease of Zhejiang Province, Zhejiang Provincial Clinical Research
Center for Mental Disorders, The Affiliated Wenzhou Kangning Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
- Monash
Institute of Pharmaceutical Sciences, Monash
University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Ali-Reza Shokouhi
- Monash
Institute of Pharmaceutical Sciences, Monash
University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Nicolas H. Voelcker
- Monash
Institute of Pharmaceutical Sciences, Monash
University, 381 Royal Parade, Parkville, VIC 3052, Australia
- Melbourne
Centre for Nanofabrication, Victorian Node of the Australian National
Fabrication Facility, 151 Wellington Road, Clayton, VIC 3168, Australia
- Department
of Materials Science and Engineering, Monash
University, 22 Alliance Lane, Clayton, VIC 3168, Australia
| | - Roey Elnathan
- Monash
Institute of Pharmaceutical Sciences, Monash
University, 381 Royal Parade, Parkville, VIC 3052, Australia
- Melbourne
Centre for Nanofabrication, Victorian Node of the Australian National
Fabrication Facility, 151 Wellington Road, Clayton, VIC 3168, Australia
- School
of Medicine, Faculty of Health, Deakin University, Waurn Ponds, VIC 3216, Australia
- Institute
for Frontier Materials, Deakin University, Geelong Waurn Ponds campus, VIC 3216, Australia
- The
Institute for Mental and Physical Health and Clinical Translation,
School of Medicine, Deakin University, Geelong Waurn Ponds Campus, Melbourne, VIC 3216, Australia
| |
Collapse
|
8
|
Chau CC, Maffeo CM, Aksimentiev A, Radford SE, Hewitt EW, Actis P. Single molecule delivery into living cells. Nat Commun 2024; 15:4403. [PMID: 38782907 PMCID: PMC11116494 DOI: 10.1038/s41467-024-48608-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 05/02/2024] [Indexed: 05/25/2024] Open
Abstract
Controlled manipulation of cultured cells by delivery of exogenous macromolecules is a cornerstone of experimental biology. Here we describe a platform that uses nanopipettes to deliver defined numbers of macromolecules into cultured cell lines and primary cells at single molecule resolution. In the nanoinjection platform, the nanopipette is used as both a scanning ion conductance microscope (SICM) probe and an injection probe. The SICM is used to position the nanopipette above the cell surface before the nanopipette is inserted into the cell into a defined location and to a predefined depth. We demonstrate that the nanoinjection platform enables the quantitative delivery of DNA, globular proteins, and protein fibrils into cells with single molecule resolution and that delivery results in a phenotypic change in the cell that depends on the identity of the molecules introduced. Using experiments and computational modeling, we also show that macromolecular crowding in the cell increases the signal-to-noise ratio for the detection of translocation events, thus the cell itself enhances the detection of the molecules delivered.
Collapse
Affiliation(s)
- Chalmers C Chau
- School of Molecular and Cellular Biology and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
- School of Electronic and Electrical Engineering and Pollard Institute, University of Leeds, Leeds, LS2 9JT, UK
- Bragg Centre for Materials Research, University of Leeds, Leeds, UK
| | - Christopher M Maffeo
- Department of Physics, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Aleksei Aksimentiev
- Department of Physics, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Sheena E Radford
- School of Molecular and Cellular Biology and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Eric W Hewitt
- School of Molecular and Cellular Biology and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK.
| | - Paolo Actis
- School of Electronic and Electrical Engineering and Pollard Institute, University of Leeds, Leeds, LS2 9JT, UK.
- Bragg Centre for Materials Research, University of Leeds, Leeds, UK.
| |
Collapse
|
9
|
Ramalingam M, Jaisankar A, Cheng L, Krishnan S, Lan L, Hassan A, Sasmazel HT, Kaji H, Deigner HP, Pedraz JL, Kim HW, Shi Z, Marrazza G. Impact of nanotechnology on conventional and artificial intelligence-based biosensing strategies for the detection of viruses. DISCOVER NANO 2023; 18:58. [PMID: 37032711 PMCID: PMC10066940 DOI: 10.1186/s11671-023-03842-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 03/28/2023] [Indexed: 04/05/2023]
Abstract
Recent years have witnessed the emergence of several viruses and other pathogens. Some of these infectious diseases have spread globally, resulting in pandemics. Although biosensors of various types have been utilized for virus detection, their limited sensitivity remains an issue. Therefore, the development of better diagnostic tools that facilitate the more efficient detection of viruses and other pathogens has become important. Nanotechnology has been recognized as a powerful tool for the detection of viruses, and it is expected to change the landscape of virus detection and analysis. Recently, nanomaterials have gained enormous attention for their value in improving biosensor performance owing to their high surface-to-volume ratio and quantum size effects. This article reviews the impact of nanotechnology on the design, development, and performance of sensors for the detection of viruses. Special attention has been paid to nanoscale materials, various types of nanobiosensors, the internet of medical things, and artificial intelligence-based viral diagnostic techniques.
Collapse
Affiliation(s)
- Murugan Ramalingam
- School of Basic Medical Sciences, Clinical Medical College & Affiliated Hospital, Chengdu University, Chengdu, 610106 China
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, 31116 Republic of Korea
- Department of Nanobiomedical Science, Dankook University, Cheonan, 31116 Republic of Korea
- BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116 Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116 Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116 South Korea
- Department of Metallurgical and Materials Engineering, Faculty of Engineering, Atilim University, 06836 Ankara, Turkey
| | - Abinaya Jaisankar
- Centre for Biomaterials, Cellular and Molecular Theranostics, School of Mechanical Engineering, Vellore Institute of Technology, Vellore, 632014 India
| | - Lijia Cheng
- School of Basic Medical Sciences, Clinical Medical College & Affiliated Hospital, Chengdu University, Chengdu, 610106 China
| | - Sasirekha Krishnan
- Centre for Biomaterials, Cellular and Molecular Theranostics, School of Mechanical Engineering, Vellore Institute of Technology, Vellore, 632014 India
| | - Liang Lan
- School of Basic Medical Sciences, Clinical Medical College & Affiliated Hospital, Chengdu University, Chengdu, 610106 China
| | - Anwarul Hassan
- Department of Mechanical and Industrial Engineering, Biomedical Research Center, Qatar University, 2713, Doha, Qatar
| | - Hilal Turkoglu Sasmazel
- Department of Metallurgical and Materials Engineering, Faculty of Engineering, Atilim University, 06836 Ankara, Turkey
| | - Hirokazu Kaji
- Department of Biomechanics, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, 101-0062 Japan
| | - Hans-Peter Deigner
- Institute of Precision Medicine, Medical and Life Sciences Faculty, Furtwangen University, 78054 Villingen-Schwenningen, Germany
| | - Jose Luis Pedraz
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country, 01006 Vitoria-Gasteiz, Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine, 28029 Madrid, Spain
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, 31116 Republic of Korea
- Department of Nanobiomedical Science, Dankook University, Cheonan, 31116 Republic of Korea
- BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116 Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116 Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116 South Korea
| | - Zheng Shi
- School of Basic Medical Sciences, Clinical Medical College & Affiliated Hospital, Chengdu University, Chengdu, 610106 China
| | - Giovanna Marrazza
- Department of Chemistry “Ugo Schiff”, University of Florence, 50019 Sesto Fiorentino, Florence, Italy
| |
Collapse
|
10
|
Kim H, Gu C, Mustfa SA, Martella DA, Wang C, Wang Y, Chiappini C. CRISPR/Cas-Assisted Nanoneedle Sensor for Adenosine Triphosphate Detection in Living Cells. ACS APPLIED MATERIALS & INTERFACES 2023; 15:49964-49973. [PMID: 37769296 PMCID: PMC10623508 DOI: 10.1021/acsami.3c07918] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 09/18/2023] [Indexed: 09/30/2023]
Abstract
The clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein (Cas) (CRISPR/Cas) systems have recently emerged as powerful molecular biosensing tools based on their collateral cleavage activity due to their simplicity, sensitivity, specificity, and broad applicability. However, the direct application of the collateral cleavage activity for in situ intracellular detection is still challenging. Here, we debut a CRISPR/Cas-assisted nanoneedle sensor (nanoCRISPR) for intracellular adenosine triphosphate (ATP), which avoids the challenges associated with intracellular collateral cleavage by introducing a two-step process of intracellular target recognition, followed by extracellular transduction and detection. ATP recognition occurs by first presenting in the cell cytosol an aptamer-locked Cas12a activator conjugated to nanoneedles; the recognition event unlocks the activator immobilized on the nanoneedles. The nanoneedles are then removed from the cells and exposed to the Cas12a/crRNA complex, where the activator triggers the cleavage of an ssDNA fluorophore-quencher pair, generating a detectable fluorescence signal. NanoCRISPR has an ATP detection limit of 246 nM and a dynamic range from 1.56 to 50 μM. Importantly, nanoCRISPR can detect intracellular ATP in 30 min in live cells without impacting cell viability. We anticipate that the nanoCRISPR approach will contribute to broadening the biomedical applications of CRISPR/Cas sensors for the detection of diverse intracellular molecules in living systems.
Collapse
Affiliation(s)
- Hongki Kim
- Centre
for Craniofacial and Regenerative Biology, King’s College London, London SE1 9RT, U.K.
- Department
of Chemistry, Kongju National University, Gongju 32588, Republic of Korea
| | - Chenlei Gu
- Centre
for Craniofacial and Regenerative Biology, King’s College London, London SE1 9RT, U.K.
- London
Centre for Nanotechnology, King’s
College London, London SE1 9RT, U.K.
| | - Salman Ahmad Mustfa
- Centre
for Craniofacial and Regenerative Biology, King’s College London, London SE1 9RT, U.K.
| | | | - Cong Wang
- Centre
for Craniofacial and Regenerative Biology, King’s College London, London SE1 9RT, U.K.
- London
Centre for Nanotechnology, King’s
College London, London SE1 9RT, U.K.
| | - Yikai Wang
- Centre
for Craniofacial and Regenerative Biology, King’s College London, London SE1 9RT, U.K.
- London
Centre for Nanotechnology, King’s
College London, London SE1 9RT, U.K.
| | - Ciro Chiappini
- Centre
for Craniofacial and Regenerative Biology, King’s College London, London SE1 9RT, U.K.
- London
Centre for Nanotechnology, King’s
College London, London SE1 9RT, U.K.
| |
Collapse
|
11
|
Chen Y, Yoh HZ, Shokouhi AR, Murayama T, Suu K, Morikawa Y, Voelcker NH, Elnathan R. Role of actin cytoskeleton in cargo delivery mediated by vertically aligned silicon nanotubes. J Nanobiotechnology 2022; 20:406. [PMID: 36076230 PMCID: PMC9461134 DOI: 10.1186/s12951-022-01618-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 08/17/2022] [Indexed: 11/10/2022] Open
Abstract
Nanofabrication technologies have been recently applied to the development of engineered nano–bio interfaces for manipulating complex cellular processes. In particular, vertically configurated nanostructures such as nanoneedles (NNs) have been adopted for a variety of biological applications such as mechanotransduction, biosensing, and intracellular delivery. Despite their success in delivering a diverse range of biomolecules into cells, the mechanisms for NN-mediated cargo transport remain to be elucidated. Recent studies have suggested that cytoskeletal elements are involved in generating a tight and functional cell–NN interface that can influence cargo delivery. In this study, by inhibiting actin dynamics using two drugs—cytochalasin D (Cyto D) and jasplakinolide (Jas), we demonstrate that the actin cytoskeleton plays an important role in mRNA delivery mediated by silicon nanotubes (SiNTs). Specifically, actin inhibition 12 h before SiNT-cellular interfacing (pre-interface treatment) significantly dampens mRNA delivery (with efficiencies dropping to 17.2% for Cyto D and 33.1% for Jas) into mouse fibroblast GPE86 cells, compared to that of untreated controls (86.9%). However, actin inhibition initiated 2 h after the establishment of GPE86 cell–SiNT interface (post-interface treatment), has negligible impact on mRNA transfection, maintaining > 80% efficiency for both Cyto D and Jas treatment groups. The results contribute to understanding potential mechanisms involved in NN-mediated intracellular delivery, providing insights into strategic design of cell–nano interfacing under temporal control for improved effectiveness.
Collapse
Affiliation(s)
- Yaping Chen
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia. .,Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia.
| | - Hao Zhe Yoh
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia.,Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia.,Commonwealth Scientific and Industrial Research Organization (CSIRO), Clayton, VIC, 3168, Australia
| | - Ali-Reza Shokouhi
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia.,Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
| | - Takahide Murayama
- Institute of Semiconductor and Electronics Technologies, ULVAC Inc, 1220-1 Suyama, Susono, Shizuoka, 410-1231, Japan
| | - Koukou Suu
- Institute of Semiconductor and Electronics Technologies, ULVAC Inc, 1220-1 Suyama, Susono, Shizuoka, 410-1231, Japan
| | - Yasuhiro Morikawa
- Institute of Semiconductor and Electronics Technologies, ULVAC Inc, 1220-1 Suyama, Susono, Shizuoka, 410-1231, Japan
| | - Nicolas H Voelcker
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia. .,Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia. .,Commonwealth Scientific and Industrial Research Organization (CSIRO), Clayton, VIC, 3168, Australia. .,Department of Materials Science and Engineering, Monash University, 22 Alliance Lane, Clayton, VIC, 3168, Australia. .,INM-Leibnitz Institute for New Materials, Campus D2 2, 66123, Saarbrücken, Germany.
| | - Roey Elnathan
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia. .,Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia. .,School of Medicine, Faculty of Health, Deakin University, Waurn Ponds, Geelong, VIC, 3216, Australia. .,Institute for Frontier Materials, Deakin University, Geelong Waurn Ponds campus, Geelong, VIC, 3216, Australia.
| |
Collapse
|