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
|
Camacho S, Aoki PHB, Ekstrand F, Oliveira ON, Prinz CN. Enhancing Photothermal Therapy Against Breast Cancer Cells by Modulating the End Point of Gold Shell-Isolated Nanoparticles Using Nanostraw-Assisted Injection. ACS APPLIED MATERIALS & INTERFACES 2025; 17:27816-27828. [PMID: 40299396 PMCID: PMC12086757 DOI: 10.1021/acsami.5c00084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 04/10/2025] [Accepted: 04/10/2025] [Indexed: 04/30/2025]
Abstract
Gold shell-isolated nanoparticles (AuSHINs) are promising photothermal therapy (PTT) agents for cancer treatment due to their excellent photoconversion efficiency, biocompatibility, colloidal stability, and tunable properties, including size, shape, and surface functionalization. However, their therapeutic efficacy in in vitro assays is often limited by poor cellular uptake and lysosomal entrapment, which can result in nanoparticle degradation and a reduction in PTT effectiveness. In this study, we demonstrate that nanostraw-assisted injection enhances the PTT efficacy of AuSHINs compared to the conventional incubation method, as evaluated in human breast cancer cell lines: adenocarcinoma cells (MDA-MB-231) and glandular carcinoma cells (MCF7). This enhancement is attributed to three differences between the delivery methods: nanoparticle internalization, intracellular targeting, and the progression of cell death pathways. Nanostraw injection resulted in approximately 10-fold higher internalization of AuSHINs compared to 0.5-h incubation. Confocal fluorescence microscopy revealed that AuSHINs delivered via conventional incubation predominantly localize within lysosomes, whereas those introduced through nanostraw-assisted injection primarily targeted the endoplasmic reticulum (ER), thus avoiding lysosomal degradation. This differential targeting led to approximately a 2-fold higher reduction in the viability of photoactivated breast cancer cells treated with nanostraw-delivered AuSHINs. Furthermore, nanostraw-assisted injection accelerated the initiation of apoptosis relative to incubation. PTT-induced cell death was more pronounced in MCF7 cells compared to MDA-MB-231 cells, reflecting the higher resistance to therapy of the latter. These findings highlight the potential of nanostraw-assisted injection to enhance PTT, and we now face the challenge of integrating it into in vivo delivery strategies.
Collapse
Affiliation(s)
- Sabrina
A. Camacho
- School
of Sciences, Humanities and Languages, São
Paulo State University (UNESP), Assis, SP 19806-900, Brazil
| | - Pedro H. B. Aoki
- School
of Sciences, Humanities and Languages, São
Paulo State University (UNESP), Assis, SP 19806-900, Brazil
| | - Frida Ekstrand
- Division
of Solid-State Physics and NanoLund, Lund
University, 221 00 Lund, Sweden
| | - Osvaldo N. Oliveira
- São
Carlos Institute of Physics, University
of São Paulo (USP), São Carlos, SP 13566-590, Brazil
| | - Christelle N. Prinz
- Division
of Solid-State Physics and NanoLund, Lund
University, 221 00 Lund, Sweden
| |
Collapse
|
3
|
Zhang G, Mu R, Ma Y, Li B. Intracellular Delivery Enabled by Squeezing Mechanoporation. SMALL METHODS 2025:e2500338. [PMID: 40357698 DOI: 10.1002/smtd.202500338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/20/2025] [Indexed: 05/15/2025]
Abstract
Squeeze mechanoporation, as an emerging method, plays an important role in intracellular delivery. It brings new opportunities to cutting-edge fields such as cell therapy, gene editing, and vaccine production, and it promises to revolutionize traditional drug delivery and treatment paradigms. By leveraging the viscoelastic properties of cells, this technique induces cell deformation under external force, creating transient micropores in cell membranes for the efficient and high-throughput delivery of diverse exogenous substances, such as nucleic acids, antibodies, nanomaterials, and drugs. This review comprehensively summarizes current advances in mechanical squeezing-mediated intracellular delivery, delving deeply into its fundamental principles, unique advantages, latest applications, optimization strategies, existing challenges, corresponding solutions, and future development directions. With the aim of highlighting the immense potential and promising prospects of these techniques in the field of biomanufacturing and cell therapy.
Collapse
Affiliation(s)
- Guorui Zhang
- State Key Laboratory of Solid Lubrication, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
| | - Rong Mu
- State Key Laboratory of Solid Lubrication, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
| | - Yanfei Ma
- State Key Laboratory of Solid Lubrication, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Shandong Laboratory of Advanced Materials and Green Manufacturing at Yantai, Shandong, 264006, China
| | - Bin Li
- State Key Laboratory of Solid Lubrication, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Shandong Laboratory of Advanced Materials and Green Manufacturing at Yantai, Shandong, 264006, China
| |
Collapse
|
4
|
Sahu SU, Castro M, Muldoon JJ, Asija K, Wyman SK, Krishnappa N, de Oñate L, Eyquem J, Nguyen DN, Wilson RC. Peptide-enabled ribonucleoprotein delivery for CRISPR engineering (PERC) in primary human immune cells and hematopoietic stem cells. Nat Protoc 2025:10.1038/s41596-025-01154-8. [PMID: 40032999 DOI: 10.1038/s41596-025-01154-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 10/10/2024] [Indexed: 03/05/2025]
Abstract
Peptide-enabled ribonucleoprotein delivery for CRISPR engineering (PERC) is a new approach for ex vivo genome editing of primary human cells. PERC uses a single amphiphilic peptide reagent to mediate intracellular delivery of the same pre-formed CRISPR ribonucleoprotein enzymes that are broadly used in research and therapeutics, resulting in high-efficiency editing of stimulated immune cells and cultured hematopoietic stem and progenitor cells (HSPCs). PERC facilitates nuclease-mediated gene knockout, precise transgene knock-in and base editing. The protocol involves mixing the CRISPR ribonucleoprotein enzyme with peptide and then incubating with cultured cells. For efficient transgene knock-in, adeno-associated virus (AAV) homology-directed repair template (HDRT) DNA may be included. In contrast to electroporation, PERC is appealing because it needs no dedicated hardware and has less impact on cell phenotype and viability. Because of the gentle nature of PERC, delivery can be performed multiple times without substantial impact to cell health or phenotype. Editing efficiencies can surpass 90% when using either Cas9 or Cas12a in primary T cells or HSPCs. After 3 h dedicated to reagent preparation, the PERC delivery step can be completed in 1 h, with the associated cell culture steps taking 3-7 d total. Because the protocol calls for only three readily available reagents (protein, RNA and peptide) and does not require dedicated hardware for any step, PERC demands no special expertise and is exceptionally straightforward to adopt. The inherent compatibility of PERC with established cell engineering pipelines makes the protocol appealing for rapid deployment in research and clinical settings.
Collapse
Affiliation(s)
- Srishti U Sahu
- Innovative Genomics Institute, University of California Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, USA
- California Institute for Quantitative Biosciences at University of California Berkeley, Berkeley, CA, USA
| | - Madalena Castro
- Innovative Genomics Institute, University of California Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, USA
- California Institute for Quantitative Biosciences at University of California Berkeley, Berkeley, CA, USA
| | - Joseph J Muldoon
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Kunica Asija
- Innovative Genomics Institute, University of California Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, USA
- California Institute for Quantitative Biosciences at University of California Berkeley, Berkeley, CA, USA
| | - Stacia K Wyman
- Innovative Genomics Institute, University of California Berkeley, Berkeley, CA, USA
| | | | - Lorena de Oñate
- Innovative Genomics Institute, University of California Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, USA
| | - Justin Eyquem
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, University of California San Francisco, San Francisco, CA, USA
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | - David N Nguyen
- Innovative Genomics Institute, University of California Berkeley, Berkeley, CA, USA
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Ross C Wilson
- Innovative Genomics Institute, University of California Berkeley, Berkeley, CA, USA.
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, USA.
- California Institute for Quantitative Biosciences at University of California Berkeley, Berkeley, CA, USA.
| |
Collapse
|
5
|
Hughes D, Mousa AH, Musumeci C, Larsson M, Shameem MA, Aydemir U, Schmiderer L, Larsson J, Berggren M, Ek F, Olsson R, Hjort M. Lithography-Free Water Stable Conductive Polymer Nanowires. NANO LETTERS 2025; 25:3059-3065. [PMID: 39943879 PMCID: PMC11869357 DOI: 10.1021/acs.nanolett.4c05016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/31/2025] [Accepted: 01/31/2025] [Indexed: 02/27/2025]
Abstract
Free-standing nanowires can gain intracellular access without causing stress or apoptosis. Current approaches to generate nanowires focus on lithographic patterning and inorganic materials (Si, GaAs, Al2O3, etc.) while organic materials are less explored. Use of organic conductive polymers allows for the creation of soft mixed ion-electron conducting nanowires. Processing conductive polymers into nanowires is challenging due to the harsh chemicals and processing conditions used. Here, we demonstrate a lithography-free and scalable method to generate all-organic, water-stable nanowires composed of conductive polymers. A nanoporous membrane is filled with conductive polymer in solution, followed by a cross-linking step to make the polymer water stable. The surface of the membrane is anisotropically etched using a reactive ion etcher to reveal the polymer inside the pores, which extends from the membrane as nanowires. We interface the nanowires with model algal cells and human primary hematopoietic stem and progenitor cells.
Collapse
Affiliation(s)
- Damien Hughes
- Chemical
Biology & Therapeutics, Department of Experimental Medical Science, Lund University, SE-221 84, Lund, Sweden
| | - Abdelrazek H. Mousa
- Department
of Chemistry and Molecular Biology, University
of Gothenburg, SE-405 30, Gothenburg, Sweden
| | - Chiara Musumeci
- Laboratory
of Organic Electronics, Department of Science and Technology, Linköping University, SE-60174, Norrkoping, Sweden
| | - Malte Larsson
- Chemical
Biology & Therapeutics, Department of Experimental Medical Science, Lund University, SE-221 84, Lund, Sweden
| | - Muhammad Anwar Shameem
- Department
of Chemistry and Molecular Biology, University
of Gothenburg, SE-405 30, Gothenburg, Sweden
| | - Umut Aydemir
- Chemical
Biology & Therapeutics, Department of Experimental Medical Science, Lund University, SE-221 84, Lund, Sweden
| | - Ludwig Schmiderer
- Division
of Molecular Medicine and Gene Therapy, Department of Laboratory Medicine
and Lund Stem Cell Center, Lund University, SE-221 00, Lund, Sweden
| | - Jonas Larsson
- Division
of Molecular Medicine and Gene Therapy, Department of Laboratory Medicine
and Lund Stem Cell Center, Lund University, SE-221 00, Lund, Sweden
| | - Magnus Berggren
- Laboratory
of Organic Electronics, Department of Science and Technology, Linköping University, SE-60174, Norrkoping, Sweden
| | - Fredrik Ek
- Chemical
Biology & Therapeutics, Department of Experimental Medical Science, Lund University, SE-221 84, Lund, Sweden
- Chemical
Biology Consortium Sweden (CBCS), Karolinska
Institute, S-171 21, Stockholm, Sweden
| | - Roger Olsson
- Chemical
Biology & Therapeutics, Department of Experimental Medical Science, Lund University, SE-221 84, Lund, Sweden
- Department
of Chemistry and Molecular Biology, University
of Gothenburg, SE-405 30, Gothenburg, Sweden
- Chemical
Biology Consortium Sweden (CBCS), Karolinska
Institute, S-171 21, Stockholm, Sweden
| | - Martin Hjort
- Chemical
Biology & Therapeutics, Department of Experimental Medical Science, Lund University, SE-221 84, Lund, Sweden
- Chemical
Biology Consortium Sweden (CBCS), Karolinska
Institute, S-171 21, Stockholm, Sweden
| |
Collapse
|
6
|
Zhao S, Gao K, Han H, Stenzel M, Yin B, Song H, Lawanprasert A, Nielsen JE, Sharma R, Arogundade OH, Pimcharoen S, Chen YJ, Paul A, Tuma J, Collins MG, Wyle Y, Cranick MG, Burgstone BW, Perez BS, Barron AE, Smith AM, Lee HY, Wang A, Murthy N. Acid-degradable lipid nanoparticles enhance the delivery of mRNA. NATURE NANOTECHNOLOGY 2024; 19:1702-1711. [PMID: 39179796 DOI: 10.1038/s41565-024-01765-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 07/19/2024] [Indexed: 08/26/2024]
Abstract
Lipid nanoparticle (LNP)-mRNA complexes are transforming medicine. However, the medical applications of LNPs are limited by their low endosomal disruption rates, high toxicity and long tissue persistence times. LNPs that rapidly hydrolyse in endosomes (RD-LNPs) could solve the problems limiting LNP-based therapeutics and dramatically expand their applications but have been challenging to synthesize. Here we present an acid-degradable linker termed 'azido-acetal' that hydrolyses in endosomes within minutes and enables the production of RD-LNPs. Acid-degradable lipids composed of polyethylene glycol lipids, anionic lipids and cationic lipids were synthesized with the azido-acetal linker and used to generate RD-LNPs, which significantly improved the performance of LNP-mRNA complexes in vitro and in vivo. Collectively, RD-LNPs delivered mRNA more efficiently to the liver, lung, spleen and brains of mice and to haematopoietic stem and progenitor cells in vitro than conventional LNPs. These experiments demonstrate that engineering LNP hydrolysis rates in vivo has great potential for expanding the medical applications of LNPs.
Collapse
Affiliation(s)
- Sheng Zhao
- Department of Bioengineering and Innovative Genomics Institute, University of California, Berkeley, CA, USA
| | - Kewa Gao
- Department of Surgery, Department of Biomedical Engineering and Institute for Pediatric Regenerative Medicine/Shriners Children's, University of California, Davis, Sacramento, CA, USA
| | - Hesong Han
- Department of Bioengineering and Innovative Genomics Institute, University of California, Berkeley, CA, USA.
| | - Michael Stenzel
- Department of Bioengineering and Innovative Genomics Institute, University of California, Berkeley, CA, USA
| | - Boyan Yin
- Department of Surgery, Department of Biomedical Engineering and Institute for Pediatric Regenerative Medicine/Shriners Children's, University of California, Davis, Sacramento, CA, USA
| | - Hengyue Song
- Department of Surgery, Department of Biomedical Engineering and Institute for Pediatric Regenerative Medicine/Shriners Children's, University of California, Davis, Sacramento, CA, USA
| | - Atip Lawanprasert
- Department of Bioengineering and Innovative Genomics Institute, University of California, Berkeley, CA, USA
| | - Josefine Eilsø Nielsen
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
- Department of Bioengineering, School of Medicine, Stanford University, Stanford, CA, USA
| | - Rohit Sharma
- Department of Bioengineering and Innovative Genomics Institute, University of California, Berkeley, CA, USA
| | - Opeyemi H Arogundade
- Department of Bioengineering and Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Sopida Pimcharoen
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Yu-Ju Chen
- Department of Cellular and Integrative Physiology, University of Texas, Health Science Center at San Antonio, San Antonio, TX, USA
| | - Abhik Paul
- Department of Cellular and Integrative Physiology, University of Texas, Health Science Center at San Antonio, San Antonio, TX, USA
| | - Jan Tuma
- Department of Cellular and Integrative Physiology, University of Texas, Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Pathophysiology, Faculty of Medicine in Pilsen, Charles University, Plzen, Czech Republic
| | - Michael G Collins
- Department of Cellular and Integrative Physiology, University of Texas, Health Science Center at San Antonio, San Antonio, TX, USA
| | - Yofiel Wyle
- Department of Surgery, Department of Biomedical Engineering and Institute for Pediatric Regenerative Medicine/Shriners Children's, University of California, Davis, Sacramento, CA, USA
| | - Matileen Grace Cranick
- Department of Surgery, Department of Biomedical Engineering and Institute for Pediatric Regenerative Medicine/Shriners Children's, University of California, Davis, Sacramento, CA, USA
| | - Benjamin W Burgstone
- Department of Bioengineering and Innovative Genomics Institute, University of California, Berkeley, CA, USA
| | - Barbara S Perez
- Department of Bioengineering and Innovative Genomics Institute, University of California, Berkeley, CA, USA
| | - Annelise E Barron
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Andrew M Smith
- Department of Bioengineering and Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Hye Young Lee
- Department of Cellular and Integrative Physiology, University of Texas, Health Science Center at San Antonio, San Antonio, TX, USA
| | - Aijun Wang
- Department of Surgery, Department of Biomedical Engineering and Institute for Pediatric Regenerative Medicine/Shriners Children's, University of California, Davis, Sacramento, CA, USA.
| | - Niren Murthy
- Department of Bioengineering and Innovative Genomics Institute, University of California, Berkeley, CA, USA.
| |
Collapse
|
7
|
Zhang R, Khare P, Banerjee P, Ivan C, Schneider S, Barbaglio F, Clise-Dwyer K, Jensen VB, Thompson E, Mendoza M, Chiorazzi N, Chen SS, Yan XJJ, Jain N, Ghia P, Caligaris-Cappio F, Mendonsa R, Kasimsetty S, Swoboda R, Bayraktar R, Wierda W, Gandhi V, Calin GA, Keating MJ, Bertilaccio MTS. The DLEU2/miR-15a/miR-16-1 cluster shapes the immune microenvironment of chronic lymphocytic leukemia. Blood Cancer J 2024; 14:168. [PMID: 39438453 PMCID: PMC11496494 DOI: 10.1038/s41408-024-01142-3] [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: 06/19/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 10/25/2024] Open
Abstract
The development and progression of chronic lymphocytic leukemia (CLL) depend on genetic abnormalities and on the immunosuppressive microenvironment. We have explored the possibility that genetic drivers might be responsible for the immune cell dysregulation that shapes the protumor microenvironment. We performed a transcriptome analysis of coding and non-coding RNAs (ncRNAs) during leukemia progression in the Rag2-/-γc-/- MEC1-based xenotransplantation model. The DLEU2/miR-16 locus was found downmodulated in monocytes/macrophages of leukemic mice. To validate the role of this cluster in the tumor immune microenvironment, we generated a mouse model that simultaneously mimics the overexpression of hTCL1 and the germline deletion of the minimal deleted region (MDR) encoding the DLEU2/miR-15a/miR-16-1 cluster. This model provides an innovative and faster CLL system where monocyte differentiation and macrophage polarization are exacerbated, and T-cells are dysfunctional. MDR deletion inversely correlates with the levels of predicted target proteins including BCL2 and PD1/PD-L1 on murine CLL cells and immune cells. The inverse correlation of miR-15a/miR-16-1 with target proteins has been confirmed on patient-derived immune cells. Forced expression of miR-16-1 interferes with monocyte differentiation into tumor-associated macrophages, indicating that selected ncRNAs drive the protumor phenotype of non-malignant immune cells.
Collapse
MESH Headings
- MicroRNAs/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Animals
- Mice
- Tumor Microenvironment/immunology
- Humans
- RNA, Long Noncoding/genetics
- Tumor Suppressor Proteins/genetics
- Multigene Family
Collapse
Affiliation(s)
- Ronghua Zhang
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Priyanka Khare
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Priyanka Banerjee
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Bryan, TX, USA
| | - Cristina Ivan
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Caris Life Sciences, Irving, TX, USA
| | - Sarah Schneider
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center, UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Federica Barbaglio
- Division of Experimental Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Karen Clise-Dwyer
- Department of Hematopoietic Biology and Malignancy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vanessa Behrana Jensen
- Department of Veterinary Medicine and Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Erika Thompson
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marisela Mendoza
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nicholas Chiorazzi
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Uniondale, NY, USA
- Departments of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Uniondale, NY, USA
- Northwell Health Cancer Institute, Lake Success, NY, USA
| | - Shih-Shih Chen
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Xiao-Jie Joy Yan
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Nitin Jain
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paolo Ghia
- B cell neoplasia Unit, Division of Experimental Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Federico Caligaris-Cappio
- Division of Experimental Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
- AIRC (Associazione Italiana per la Ricerca sul Cancro), 20123, Milan, Italy
| | | | | | | | - Recep Bayraktar
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - William Wierda
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Varsha Gandhi
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - George A Calin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael J Keating
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | |
Collapse
|
8
|
Sahu S, Castro M, Muldoon JJ, Asija K, Wyman SK, Krishnappa N, de Onate L, Eyquem J, Nguyen DN, Wilson RC. Peptide-enabled ribonucleoprotein delivery for CRISPR engineering (PERC) in primary human immune cells and hematopoietic stem cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.14.603391. [PMID: 39071446 PMCID: PMC11275745 DOI: 10.1101/2024.07.14.603391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Peptide-enabled ribonucleoprotein delivery for CRISPR engineering (PERC) is a new approach for ex vivo genome editing of primary human cells. PERC uses a single amphiphilic peptide reagent to mediate intracellular delivery of the same pre-formed CRISPR ribonucleoprotein enzymes that are broadly used in research and therapeutics, resulting in high-efficiency editing of stimulated immune cells and cultured hematopoietic stem and progenitor cells (HSPCs). PERC facilitates nuclease-mediated gene knockout, precise transgene knock-in, and base editing. PERC involves mixing the CRISPR ribonucleoprotein enzyme with peptide and then incubating the formulation with cultured cells. For efficient transgene knock-in, adeno-associated virus (AAV) bearing homology-directed repair template DNA may be included. In contrast to electroporation, PERC is appealing as it requires no dedicated hardware and has less impact on cell phenotype and viability. Due to the gentle nature of PERC, delivery can be performed multiple times without substantial impact to cell health or phenotype. Here we report methods for improved PERC-mediated editing of T cells as well as novel methods for PERC-mediated editing of HSPCs, including knockout and precise knock-in. Editing efficiencies can surpass 90% using either Cas9 or Cas12a in primary T cells or HSPCs. Because PERC calls for only three readily available reagents - protein, RNA, and peptide - and does not require dedicated hardware for any step, PERC demands no special expertise and is exceptionally straightforward to adopt. The inherent compatibility of PERC with established cell engineering pipelines makes this approach appealing for rapid deployment in research and clinical settings.
Collapse
|
9
|
Hu T, Kumar AR, Luo Y, Tay A. Automating CAR-T Transfection with Micro and Nano-Technologies. SMALL METHODS 2024; 8:e2301300. [PMID: 38054597 DOI: 10.1002/smtd.202301300] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/15/2023] [Indexed: 12/07/2023]
Abstract
Cancer poses a significant health challenge, with traditional treatments like surgery, radiotherapy, and chemotherapy often lacking in cell specificity and long-term curative potential. Chimeric antigen receptor T cell (CAR-T) therapy,utilizing genetically engineered T cells to target cancer cells, is a promising alternative. However, its high cost limits widespread application. CAR-T manufacturing process encompasses three stages: cell isolation and activation, transfection, and expansion.While the first and last stages have straightforward, commercially available automation technologies, the transfection stage lags behind. Current automated transfection relies on viral vectors or bulk electroporation, which have drawbacks such as limited cargo capacity and significant cell disturbance. Conversely, micro and nano-tool methods offer higher throughput and cargo flexibility, yet their automation remains underexplored.In this perspective, the progress in micro and nano-engineering tools for CAR-T transfection followed by a discussion to automate them is described. It is anticipated that this work can inspire the community working on micro and nano transfection techniques to examine how their protocols can be automated to align with the growing interest in automating CAR-T manufacturing.
Collapse
Affiliation(s)
- Tianmu Hu
- Engineering Science Programme, National University of Singapore, Singapore, 117575, Singapore
| | - Arun Rk Kumar
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Yikai Luo
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
| | - Andy Tay
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
- Tissue Engineering Programme, National University of Singapore, Singapore, 117510, Singapore
| |
Collapse
|
10
|
Ekstrand F, Mapar M, Ruhrmann S, Bacos K, Ling C, Prinz CN. Achieving efficient clonal beta cells transfection using nanostraw/nanopore-assisted electroporation. RSC Adv 2024; 14:22244-22252. [PMID: 39010923 PMCID: PMC11247384 DOI: 10.1039/d4ra02791d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 07/08/2024] [Indexed: 07/17/2024] Open
Abstract
The prospect of being able to efficiently inject large plasmids in insulin-producing beta cells is very attractive for diabetes research. However, conventional transfection methods suffer from high cytotoxicity or low transfection efficiency, which negatively affect their outcome. In contrast, nanostraw electroporation is a gentle method that can provide a high transfection efficiency while maintaining high cell viability. While nanostraw electroporation has gone through some method optimization in the past, such as tuning the pulse frequency, amplitude, and duration, the effect of other parameters has not been thoroughly investigated. Here, we demonstrate efficient transfection of clonal beta cells and investigate the effect of voltage at a fixed inter-electrode distance, cell density, and cargo solution conductivity on transfection efficiency. We used GFP-encoding DNA plasmids stained with an intercalating dye to enable immediate analysis and assessment of the electrophoretic transport of cargo. Moreover, we ran simulations to assess how cargo buffer conductivity impacts the transfection efficiency by affecting the voltage drop on the nanostraws and cell membrane during electroporation. Both experiments and simulations show that MilliQ water as the cargo buffer yields the best transfection efficiency. We also show that the cell density should be adjusted to maximize the number of cells interfacing the nanostraws and avoid cell stacking. Finally, we compared the transfection efficiency when using nanostraws and nanopores. Whereas the amount of GFP plasmids injected using nanostraws is larger than for nanopores, the outcome in terms of GFP fluorescence 48 h after transfection was worse than for nanopores. Moreover, when using nanostraws, fewer cells were found on the substrate 48 h after transfection compared to when using nanopores. This suggests that injecting substantial amounts of plasmids in cells can affect their proliferation and/or viability, and that nanopore electroporation, as a simpler method, is an interesting alternative to nanostraws in achieving efficient and gentle clonal beta cell transfection.
Collapse
Affiliation(s)
- Frida Ekstrand
- Division of Solid State Physics, NanoLund, Lund University 221 00 Lund Sweden
| | - Mokhtar Mapar
- Division of Solid State Physics, NanoLund, Lund University 221 00 Lund Sweden
| | - Sabrina Ruhrmann
- Epigenetics and Diabetes Unit, Lund University Diabetes Centre, Department of Clinical Sciences, Scania University Hospital 214 28 Malmö Sweden
| | - Karl Bacos
- Epigenetics and Diabetes Unit, Lund University Diabetes Centre, Department of Clinical Sciences, Scania University Hospital 214 28 Malmö Sweden
| | - Charlotte Ling
- Epigenetics and Diabetes Unit, Lund University Diabetes Centre, Department of Clinical Sciences, Scania University Hospital 214 28 Malmö Sweden
| | - Christelle N Prinz
- Division of Solid State Physics, NanoLund, Lund University 221 00 Lund Sweden
| |
Collapse
|
11
|
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
|
12
|
Liu F, Su R, Jiang X, Wang S, Mu W, Chang L. Advanced micro/nano-electroporation for gene therapy: recent advances and future outlook. NANOSCALE 2024; 16:10500-10521. [PMID: 38757536 DOI: 10.1039/d4nr01408a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
Gene therapy is a promising disease treatment approach by editing target genes, and thus plays a fundamental role in precision medicine. To ensure gene therapy efficacy, the effective delivery of therapeutic genes into specific cells is a key challenge. Electroporation utilizes short electric pulses to physically break the cell membrane barrier, allowing gene transfer into the cells. It dodges the off-target risks associated with viral vectors, and also stands out from other physical-based gene delivery methods with its high-throughput and cargo-accelerating features. In recent years, with the help of advanced micro/nanotechnology, micro/nanostructure-integrated electroporation (micro/nano-electroporation) techniques and devices have significantly improved cell viability, transfection efficiency and dose controllability of the electroporation strategy, enhancing its application practicality especially in vivo. This technical advancement makes micro/nano-electroporation an effective and versatile tool for gene therapy. In this review, we first introduce the evolution of electroporation technique with a brief explanation of the perforation mechanism, and then provide an overview of the recent advancements and prospects of micro/nano-electroporation technology in the field of gene therapy. To comprehensively showcase the latest developments of micro/nano-electroporation technology in gene therapy, we focus on discussing micro/nano-electroporation devices and current applications at both in vitro and in vivo levels. Additionally, we outline the ongoing clinical studies of gene electrotransfer (GET), revealing the tremendous potential of electroporation-based gene delivery in disease treatment and healthcare. Lastly, the challenges and future directions in this field are discussed.
Collapse
Affiliation(s)
- Feng Liu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Rongtai Su
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Xinran Jiang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Siqi Wang
- Department of General Surgery and Obesity and Metabolic Disease Center, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Wei Mu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
- School of Engineering Medicine, Beihang University, Beijing, 100191, China
- Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of the People's Republic of China, Beijing, 100191, China
| | - Lingqian Chang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| |
Collapse
|
13
|
Muralidharan A, Boukany PE. Electrotransfer for nucleic acid and protein delivery. Trends Biotechnol 2024; 42:780-798. [PMID: 38102019 DOI: 10.1016/j.tibtech.2023.11.009] [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: 10/19/2023] [Revised: 11/15/2023] [Accepted: 11/15/2023] [Indexed: 12/17/2023]
Abstract
Electrotransfer of nucleic acids and proteins has become crucial in biotechnology for gene augmentation and genome editing. This review explores the applications of electrotransfer in both ex vivo and in vivo scenarios, emphasizing biomedical uses. We provide insights into completed clinical trials and successful instances of nucleic acid and protein electrotransfer into therapeutically relevant cells such as immune cells and stem and progenitor cells. In addition, we delve into emerging areas of electrotransfer where nanotechnology and deep learning techniques overcome the limitations of traditional electroporation.
Collapse
Affiliation(s)
- Aswin Muralidharan
- Department of Bionanoscience, Delft University of Technology, van der Maasweg 9, 2629 HZ Delft, The Netherlands; Kavli Institute of Nanoscience, Delft University of Technology, van der Maasweg 9, 2629 HZ Delft, The Netherlands.
| | - Pouyan E Boukany
- Department of Chemical Engineering, Delft University of Technology, van der Maasweg 9, 2629 HZ Delft, The Netherlands.
| |
Collapse
|
14
|
Patange S, Maragh S. Fire Burn and Cauldron Bubble: What Is in Your Genome Editing Brew? Biochemistry 2023; 62:3500-3511. [PMID: 36306429 PMCID: PMC10734218 DOI: 10.1021/acs.biochem.2c00431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 09/28/2022] [Indexed: 11/28/2022]
Abstract
Genome editing is a rapidly evolving biotechnology with the potential to transform many sectors of industry such as agriculture, biomanufacturing, and medicine. This technology is enabled by an ever-growing portfolio of biomolecular reagents that span the central dogma, from DNA to RNA to protein. In this paper, we draw from our unique perspective as the National Metrology Institute of the United States to bring attention to the importance of understanding and reporting genome editing formulations accurately and promoting concepts to verify successful delivery into cells. Achieving the correct understanding may be hindered by the way units, quantities, and stoichiometries are reported in the field. We highlight the variability in how editing formulations are reported in the literature and examine how a reference molecule could be used to verify the delivery of a reagent into cells. We provide recommendations on how more accurate reporting of editing formulations and more careful verification of the steps in an editing experiment can help set baseline expectations of reagent performance, toward the aim of enabling genome editing studies to be more reproducible. We conclude with a future outlook on technologies that can further our control and enable our understanding of genome editing outcomes at the single-cell level.
Collapse
Affiliation(s)
- Simona Patange
- Biosystems and Biomaterials
Division, Material Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
| | - Samantha Maragh
- Biosystems and Biomaterials
Division, Material Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
| |
Collapse
|
15
|
Liu Y, Schmiderer L, Hjort M, Lang S, Bremborg T, Rydström A, Schambach A, Larsson J, Karlsson S. Engineered human Diamond-Blackfan anemia disease model confirms therapeutic effects of clinically applicable lentiviral vector at single-cell resolution. Haematologica 2023; 108:3095-3109. [PMID: 37199130 PMCID: PMC10620578 DOI: 10.3324/haematol.2022.282068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 05/11/2023] [Indexed: 05/19/2023] Open
Abstract
Diamond-Blackfan anemia is a rare genetic bone marrow failure disorder which is usually caused by mutations in ribosomal protein genes. In the present study, we generated a traceable RPS19-deficient cell model using CRISPR-Cas9 and homology-directed repair to investigate the therapeutic effects of a clinically applicable lentiviral vector at single-cell resolution. We developed a gentle nanostraw delivery platform to edit the RPS19 gene in primary human cord bloodderived CD34+ hematopoietic stem and progenitor cells. The edited cells showed expected impaired erythroid differentiation phenotype, and a specific erythroid progenitor with abnormal cell cycle status accompanied by enrichment of TNFα/NF-κB and p53 signaling pathways was identified by single-cell RNA sequencing analysis. The therapeutic vector could rescue the abnormal erythropoiesis by activating cell cycle-related signaling pathways and promoted red blood cell production. Overall, these results establish nanostraws as a gentle option for CRISPR-Cas9- based gene editing in sensitive primary hematopoietic stem and progenitor cells, and provide support for future clinical investigations of the lentiviral gene therapy strategy.
Collapse
Affiliation(s)
- Yang Liu
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund 22184, Sweden; Department of Medicine, Huddinge, Karolinska Institutet, 14157 Stockholm.
| | - Ludwig Schmiderer
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund 22184
| | - Martin Hjort
- Chemical Biology and Therapeutics, Department of Experimental Medical Science, Lund University, 22100 Lund, Sweden; Navan Technologies, MBC Biolabs, San Carlos, CA 94070; NanoLund, Lund University, Box 118, 22100 Lund
| | - Stefan Lang
- Division of Molecular Hematology and Stem Cell Center, Lund University, Lund
| | - Tyra Bremborg
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund 22184
| | - Anna Rydström
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund 22184
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover 30625, Germany; Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School
| | - Jonas Larsson
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund 22184
| | - Stefan Karlsson
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund 22184.
| |
Collapse
|
16
|
Shokouhi AR, Chen Y, Yoh HZ, Brenker J, Alan T, Murayama T, Suu K, Morikawa Y, Voelcker NH, Elnathan R. Engineering Efficient CAR-T Cells via Electroactive Nanoinjection. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2304122. [PMID: 37434421 DOI: 10.1002/adma.202304122] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 07/10/2023] [Accepted: 07/10/2023] [Indexed: 07/13/2023]
Abstract
Chimeric antigen receptor (CAR)-T cell therapy has emerged as a promising cell-based immunotherapy approach for treating blood disorders and cancers, but genetically engineering CAR-T cells is challenging due to primary T cells' sensitivity to conventional gene delivery approaches. The current viral-based method can typically involve significant operating costs and biosafety hurdles, while bulk electroporation (BEP) can lead to poor cell viability and functionality. Here, a non-viral electroactive nanoinjection (ENI) platform is developed to efficiently negotiate the plasma membrane of primary human T cells via vertically configured electroactive nanotubes, enabling efficient delivery (68.7%) and expression (43.3%) of CAR genes in the T cells, with minimal cellular perturbation (>90% cell viability). Compared to conventional BEP, the ENI platform achieves an almost threefold higher CAR transfection efficiency, indicated by the significantly higher reporter GFP expression (43.3% compared to 16.3%). By co-culturing with target lymphoma Raji cells, the ENI-transfected CAR-T cells' ability to effectively suppress lymphoma cell growth (86.9% cytotoxicity) is proved. Taken together, the results demonstrate the platform's remarkable capacity to generate functional and effective anti-lymphoma CAR-T cells. Given the growing potential of cell-based immunotherapies, such a platform holds great promise for ex vivo cell engineering, especially in CAR-T cell therapy.
Collapse
Affiliation(s)
- 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
| | - 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
| | - Jason Brenker
- Dynamic Micro Devices (DMD) Lab, Department of Mechanical & Aerospace Engineering, Monash University, 17 College Walk, Clayton, VIC, 3168, Australia
| | - Tuncay Alan
- Dynamic Micro Devices (DMD) Lab, Department of Mechanical & Aerospace Engineering, Monash University, 17 College Walk, 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
- Department of Materials Science and Engineering, Monash University, 22 Alliance Lane, Clayton, VIC, 3168, Australia
| | - Roey Elnathan
- School of Medicine, Faculty of Health, Deakin University, Waurn Ponds, VIC, 3216, Australia
- Institute for Frontier Materials, Deakin University, Geelong Waurn Ponds campus, Waurn Ponds, 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
|
17
|
Liu J, Jiang J, Deng C, Huang X, Huang S, Liu Z, Yang J, Mo J, Chen HJ, Wang J, Xie X. Nanochannel Electro-Injection as a Versatile Platform for Efficient RNA/DNA Programming on Dendritic Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2303088. [PMID: 37381646 DOI: 10.1002/smll.202303088] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/03/2023] [Indexed: 06/30/2023]
Abstract
The utilization of dendritic cell (DC) vaccines is a promising approach in cancer immunotherapy, and the modification of DCs for the expression of tumor-associated antigens is critical for successful cancer immunotherapy. A safe and efficient method for delivering DNA/RNA into DCs without inducing maturation is beneficial to achieve successful DC transformation for cell vaccine applications, yet remains challenging. This work presents a nanochannel electro-injection (NEI) system for the safe and efficient delivery of a variety of nucleic acid molecules into DCs. The device is based on track-etched nanochannel membrane as key components, where the nano-sized channels localize the electric field on the cell membrane, enabling lower voltage (<30 V) for cell electroporation. The pulse conditions of NEI are examined so that the transfection efficiency (>70%) and biosafety (viability >85%) on delivering fluorescent dyes, plasmid DNA, messenger RNA, and circular RNA (circRNA) into DC2.4 are optimized. Primary mouse bone marrow DC can also be transfected with circRNA with 68.3% efficiency, but without remarkably affecting cellular viability or inducing DC maturation. These results suggest that NEI can be a safe and efficient transfection platform for in vitro transformation of DCs and possesses a promising potential for developing DC vaccines against cancer.
Collapse
Affiliation(s)
- Jing Liu
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, P. R. China
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Juan Jiang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, P. R. China
| | - Caiguanxi Deng
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, P. R. China
| | - Xinshuo Huang
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Shuang Huang
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Zhengjie Liu
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Jiang Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Jingshan Mo
- School of Electronic and Information Engineering, Guangdong Ocean University, Zhanjiang, 524088, P. R. China
| | - Hui-Jiuan Chen
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Ji Wang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, P. R. China
| | - Xi Xie
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, P. R. China
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| |
Collapse
|
18
|
Murugesan R, Karuppusamy KV, Marepally S, Thangavel S. Current approaches and potential challenges in the delivery of gene editing cargos into hematopoietic stem and progenitor cells. Front Genome Ed 2023; 5:1148693. [PMID: 37780116 PMCID: PMC10540692 DOI: 10.3389/fgeed.2023.1148693] [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: 01/20/2023] [Accepted: 08/17/2023] [Indexed: 10/03/2023] Open
Abstract
Advancements in gene delivery and editing have expanded the applications of autologous hematopoietic stem and progenitor cells (HSPCs) for the treatment of monogenic and acquired diseases. The gene editing toolbox is growing, and the ability to achieve gene editing with mRNA or protein delivered intracellularly by vehicles, such as electroporation and nanoparticles, has highlighted the potential of gene editing in HSPCs. Ongoing phase I/II clinical trials with gene-edited HSPCs for β-hemoglobinopathies provide hope for treating monogenic diseases. The development of safe and efficient gene editing reagents and their delivery into hard-to-transfect HSPCs have been critical drivers in the rapid translation of HSPC gene editing into clinical studies. This review article summarizes the available payloads and delivery vehicles for gene editing HSPCs and their potential impact on therapeutic applications.
Collapse
Affiliation(s)
- Ramya Murugesan
- Centre for Stem Cell Research (CSCR), A Unit of InStem Bengaluru, Christian Medical College Campus, Vellore, Tamil Nadu, India
- Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Karthik V. Karuppusamy
- Centre for Stem Cell Research (CSCR), A Unit of InStem Bengaluru, Christian Medical College Campus, Vellore, Tamil Nadu, India
- Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Srujan Marepally
- Centre for Stem Cell Research (CSCR), A Unit of InStem Bengaluru, Christian Medical College Campus, Vellore, Tamil Nadu, India
| | - Saravanabhavan Thangavel
- Centre for Stem Cell Research (CSCR), A Unit of InStem Bengaluru, Christian Medical College Campus, Vellore, Tamil Nadu, India
| |
Collapse
|
19
|
Shokouhi AR, Chen Y, Yoh HZ, Murayama T, Suu K, Morikawa Y, Brenker J, Alan T, Voelcker NH, Elnathan R. Electroactive nanoinjection platform for intracellular delivery and gene silencing. J Nanobiotechnology 2023; 21:273. [PMID: 37592297 PMCID: PMC10433684 DOI: 10.1186/s12951-023-02056-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 08/07/2023] [Indexed: 08/19/2023] Open
Abstract
BACKGROUND Nanoinjection-the process of intracellular delivery using vertically configured nanostructures-is a physical route that efficiently negotiates the plasma membrane, with minimal perturbation and toxicity to the cells. Nanoinjection, as a physical membrane-disruption-mediated approach, overcomes challenges associated with conventional carrier-mediated approaches such as safety issues (with viral carriers), genotoxicity, limited packaging capacity, low levels of endosomal escape, and poor versatility for cell and cargo types. Yet, despite the implementation of nanoinjection tools and their assisted analogues in diverse cellular manipulations, there are still substantial challenges in harnessing these platforms to gain access into cell interiors with much greater precision without damaging the cell's intricate structure. Here, we propose a non-viral, low-voltage, and reusable electroactive nanoinjection (ENI) platform based on vertically configured conductive nanotubes (NTs) that allows for rapid influx of targeted biomolecular cargos into the intracellular environment, and for successful gene silencing. The localization of electric fields at the tight interface between conductive NTs and the cell membrane drastically lowers the voltage required for cargo delivery into the cells, from kilovolts (for bulk electroporation) to only ≤ 10 V; this enhances the fine control over membrane disruption and mitigates the problem of high cell mortality experienced by conventional electroporation. RESULTS Through both theoretical simulations and experiments, we demonstrate the capability of the ENI platform to locally perforate GPE-86 mouse fibroblast cells and efficiently inject a diverse range of membrane-impermeable biomolecules with efficacy of 62.5% (antibody), 55.5% (mRNA), and 51.8% (plasmid DNA), with minimal impact on cells' viability post nanoscale-EP (> 90%). We also show gene silencing through the delivery of siRNA that targets TRIOBP, yielding gene knockdown efficiency of 41.3%. CONCLUSIONS We anticipate that our non-viral and low-voltage ENI platform is set to offer a new safe path to intracellular delivery with broader selection of cargo and cell types, and will open opportunities for advanced ex vivo cell engineering and gene silencing.
Collapse
Affiliation(s)
- 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
| | - 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
| | - 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
| | - Jason Brenker
- Department of Mechanical and Aerospace Engineering, Monash University, Wellington Rd, Clayton, VIC, 3168, Australia
| | - Tuncay Alan
- Department of Mechanical and Aerospace Engineering, Monash University, Wellington Rd, Clayton, VIC, 3168, 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.
- INM-Leibniz Institute for New Materials, Campus D2 2, 66123, Saarbrücken, Germany.
- 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.
- Faculty of Health, School of Medicine, Deakin University, Waurn Ponds, Melbourne, VIC, 3216, Australia.
- Institute for Frontier Materials, Deakin University, Geelong Waurn Ponds campus, Melbourne, 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
|
20
|
Campelo SN, Huang PH, Buie CR, Davalos RV. Recent Advancements in Electroporation Technologies: From Bench to Clinic. Annu Rev Biomed Eng 2023; 25:77-100. [PMID: 36854260 PMCID: PMC11633374 DOI: 10.1146/annurev-bioeng-110220-023800] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
Over the past decade, the increased adoption of electroporation-based technologies has led to an expansion of clinical research initiatives. Electroporation has been utilized in molecular biology for mammalian and bacterial transfection; for food sanitation; and in therapeutic settings to increase drug uptake, for gene therapy, and to eliminate cancerous tissues. We begin this article by discussing the biophysics required for understanding the concepts behind the cell permeation phenomenon that is electroporation. We then review nano- and microscale single-cell electroporation technologies before scaling up to emerging in vivo applications.
Collapse
Affiliation(s)
- Sabrina N Campelo
- Department of Biomedical Engineering and Mechanics, Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Virginia Tech, Blacksburg, Virginia, USA;
| | - Po-Hsun Huang
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Cullen R Buie
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Rafael V Davalos
- Department of Biomedical Engineering and Mechanics, Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Virginia Tech, Blacksburg, Virginia, USA;
| |
Collapse
|
21
|
Yoh HZ, Chen Y, Shokouhi AR, Thissen H, Voelcker NH, Elnathan R. The influence of dysfunctional actin on polystyrene-nanotube-mediated mRNA nanoinjection into mammalian cells. NANOSCALE 2023; 15:7737-7744. [PMID: 37066984 DOI: 10.1039/d3nr01111a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
The advancement of nanofabrication technologies has transformed the landscape of engineered nano-bio interfaces, especially with vertically aligned nanoneedles (NNs). This enables scientists to venture into new territories, widening NN applications into increasingly more complex cellular manipulation and interrogation. Specifically, for intracellular delivery application, NNs have been shown to mediate the delivery of various bioactive cargos into a wide range of cells-a physical method termed "nanoinjection". Silicon (Si) nanostructures demonstrated great potential in nanoinjection, whereas the use of polymeric NNs for nanoinjection has rarely been explored. Furthermore, the underlying mechanism of interaction at the cell-NN interface is subtle and multifaceted, and not fully understood-underpinned by the design versatility of the NN biointerface. Recent studies have suggested that actin dynamic plays a pivotal role influencing the delivery efficacy. In this study, we fabricated a new class of NNs-a programmable polymeric nanotubes (NTs)-from polystyrene (PS) cell cultureware, designed to facilitate mRNA delivery into mouse embryonic fibroblast GPE86 cells. The PSNT delivery platform was able to mediate mRNA delivery with high delivery efficiency (∼83%). We also investigated the role of actin cytoskeleton in PSNTs mediated intracellular delivery by introducing two actin inhibitors-cytochalasin D (Cyto D) and jasplakinolide (Jas)-to cause dysfunctional cytoskeleton, via inhibiting actin polymerization and depolymerization, respectively (before and after the establishment of cell-PSNT interface). By inhibiting actin dynamics 12 h before cell-PSNT interfacing (pre-interface treatment), the mRNA delivery efficiencies were significantly reduced to ∼3% for Cyto D-treated samples and ∼1% for Jas-treated sample, as compared to their post-interface (2 h after cell-PSNT interfacing) counterpart (∼46% and ∼68%, respectively). The added flexibility of PSNTs have shown to help withstand mechanical breakage stemming from cytoskeletal forces in contrast to the SiNTs. Such findings will step-change our capacity to use programmable polymeric NTs in fundamental cellular processes related to intracellular delivery.
Collapse
Affiliation(s)
- 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
| | - Yaping Chen
- 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.
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC 3168, Australia
| | - Helmut Thissen
- Commonwealth Scientific and Industrial Research Organization (CSIRO), Clayton, VIC 3168, 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, Geelong, VIC 3216, Australia
- Institute for Frontier Materials, Deakin University, Geelong Waurn Ponds Campus, Geelong, VIC 3216, Australia
- Institute for Innovation in Mental and Physical Health and Clinical Translation (IMPACT), Geelong Waurn Ponds Campus, Geelong, VIC 3216, Australia
| |
Collapse
|
22
|
Pathak N, Patino CA, Ramani N, Mukherjee P, Samanta D, Ebrahimi SB, Mirkin CA, Espinosa HD. Cellular Delivery of Large Functional Proteins and Protein-Nucleic Acid Constructs via Localized Electroporation. NANO LETTERS 2023; 23:3653-3660. [PMID: 36848135 PMCID: PMC10433461 DOI: 10.1021/acs.nanolett.2c04374] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Delivery of proteins and protein-nucleic acid constructs into live cells enables a wide range of applications from gene editing to cell-based therapies and intracellular sensing. However, electroporation-based protein delivery remains challenging due to the large sizes of proteins, their low surface charge, and susceptibility to conformational changes that result in loss of function. Here, we use a nanochannel-based localized electroporation platform with multiplexing capabilities to optimize the intracellular delivery of large proteins (β-galactosidase, 472 kDa, 75.38% efficiency), protein-nucleic acid conjugates (protein spherical nucleic acids (ProSNA), 668 kDa, 80.25% efficiency), and Cas9-ribonucleoprotein complex (160 kDa, ∼60% knock-out and ∼24% knock-in) while retaining functionality post-delivery. Importantly, we delivered the largest protein to date using a localized electroporation platform and showed a nearly 2-fold improvement in gene editing efficiencies compared to previous reports. Furthermore, using confocal microscopy, we observed enhanced cytosolic delivery of ProSNAs, which may expand opportunities for detection and therapy.
Collapse
Affiliation(s)
- Nibir Pathak
- Department of Mechanical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
- Theoretical and Applied Mechanics Program, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Cesar A Patino
- Department of Mechanical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Namrata Ramani
- Department of Materials Science and Engineering and International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Prithvijit Mukherjee
- Department of Mechanical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
- Theoretical and Applied Mechanics Program, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Devleena Samanta
- Department of Chemistry and International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Sasha B Ebrahimi
- Department of Chemical and Biological Engineering and International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Chad A Mirkin
- Department of Chemistry and International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Horacio D Espinosa
- Department of Mechanical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
- Theoretical and Applied Mechanics Program, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| |
Collapse
|
23
|
Zhang G, Kang D, Zhang Z, Li Y, Jiang J, Tu Q, Du J, Wang J. Verification and Analysis of Filter Paper-Based Intracellular Delivery of Exogenous Substances. Anal Chem 2023; 95:4353-4361. [PMID: 36623324 DOI: 10.1021/acs.analchem.2c04675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The intracellular delivery of exogenous substances is an essential technical means in the field of biomedical research, including cell therapy and gene editing. Although many delivery technologies and strategies are present, each technique has its own limitations. The delivery cost is usually a major limiting factor for general laboratories. In addition, simplifying the operation process and shortening the delivery time are key challenges. Here, we develop a filter paper-syringe (FPS) delivery method, a new type of cell permeation approach based on filter paper. The cells in a syringe are forced to pass through the filter paper quickly. During this process, external pressure forces the cells to collide and squeeze with the fiber matrix of the filter paper, causing the cells to deform rapidly, thereby enhancing the permeability of the cell membrane and realizing the delivery of exogenous substances. Moreover, the large gap between the fiber networks of filter paper can prevent the cells from bearing high pressure, thus maintaining high cell vitality. Results showed that the slow-speed filter paper used can realize efficient intracellular delivery of various exogenous substances, especially small molecular substances (e.g., 3-5 kDa dextran and siRNA). Meanwhile, we found that the FPS method not only does not require a lengthy operating step compared with the widely used liposomal delivery of siRNA but also that the delivery efficiency is similar. In conclusion, the FPS approach is a simple, easy-to-operate, and fast (about 2 s) delivery method and may be an attractive alternative to membrane destruction-based transfection.
Collapse
Affiliation(s)
- Guorui Zhang
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Di Kang
- State Key Laboratory of Veterinary Etiological Biology, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730046, P. R. China
| | - Zhonghui Zhang
- State Key Laboratory of Veterinary Etiological Biology, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730046, P. R. China
| | - Yuanchang Li
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Jingjing Jiang
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Qin Tu
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Junzheng Du
- State Key Laboratory of Veterinary Etiological Biology, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730046, P. R. China
| | - Jinyi Wang
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| |
Collapse
|
24
|
Geoghegan N, O'Loughlin M, Delaney C, Rochfort KD, Kennedy M, Kolagatla S, Podhorska L, Rodriguez BJ, Florea L, Kelleher SM. Controlled degradation of polycaprolactone-based micropillar arrays. Biomater Sci 2023; 11:3077-3091. [PMID: 36876330 PMCID: PMC10152922 DOI: 10.1039/d3bm00165b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
Herein we demonstrate the fabrication of arrays of micropillars, achieved through the combination of direct laser writing and nanoimprint lithography. By combining two diacrylate monomers, polycaprolactone dimethacrylate (PCLDMA) and 1,6-hexanediol diacrylate (HDDA), two copolymer formulations that, owing to the varying ratios of the hydrolysable ester functionalities present in the polycaprolactone moiety, can be degraded in the presence of base in a controllable manner. As such, the degradation of the micropillars can be tuned over several days as a function of PCLDMA concentration within the copolymer formulations, and the topography greatly varied over a short space of time, as visualised using scanning electron microscopy and atomic force microscopy. Crosslinked neat HDDA was used as a control material, demonstrating that the presence of the PCL was responsible for the ability of the microstructures to degrade in the controlled manner. In addition, the mass loss of the crosslinked materials was minimal, demonstrating the degradation of microstructured surfaces without loss of bulk properties was possible. Moreover, the compatibility of these crosslinked materials with mammalian cells was explored. The influence of both indirect and direct contact of the materials with A549 cells was assessed by profiling indices reflective of cytotoxicity such as morphology, adhesion, metabolic activity, oxidative balance, and release of injury markers. No significant changes in the aforementioned profile were observed in the cells cultured under these conditions for up to 72 h, with the cell-material interaction suggesting these materials may have potential in microfabrication contexts towards biomedical application purposes.
Collapse
Affiliation(s)
- Niamh Geoghegan
- School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland. .,CURAM, Science Foundation Ireland Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Mark O'Loughlin
- School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Colm Delaney
- School of Chemistry and AMBER, the SFI Research Centre for Advanced Materials and BioEngineering Research, Trinity College Dublin, the University of Dublin, College Green, Dublin 2, Ireland
| | - Keith D Rochfort
- School of Nursing, Psychotherapy, and Community Health, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Meabh Kennedy
- School of Chemical Sciences, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Srikanth Kolagatla
- School of Chemistry and AMBER, the SFI Research Centre for Advanced Materials and BioEngineering Research, Trinity College Dublin, the University of Dublin, College Green, Dublin 2, Ireland.,Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Lucia Podhorska
- School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Brian J Rodriguez
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Larisa Florea
- School of Chemistry and AMBER, the SFI Research Centre for Advanced Materials and BioEngineering Research, Trinity College Dublin, the University of Dublin, College Green, Dublin 2, Ireland
| | - Susan M Kelleher
- School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland. .,CURAM, Science Foundation Ireland Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland.,School of Chemical Sciences, Dublin City University, Glasnevin, Dublin 9, Ireland
| |
Collapse
|
25
|
Mukherjee P, Peng CY, McGuire T, Hwang JW, Puritz CH, Pathak N, Patino CA, Braun R, Kessler JA, Espinosa HD. Single cell transcriptomics reveals reduced stress response in stem cells manipulated using localized electric fields. Mater Today Bio 2023; 19:100601. [PMID: 37063248 PMCID: PMC10102005 DOI: 10.1016/j.mtbio.2023.100601] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 02/11/2023] [Accepted: 03/03/2023] [Indexed: 03/07/2023] Open
Abstract
Membrane disruption using Bulk Electroporation (BEP) is a widely used non-viral method for delivering biomolecules into cells. Recently, its microfluidic counterpart, Localized Electroporation (LEP), has been successfully used for several applications ranging from reprogramming and engineering cells for therapeutic purposes to non-destructive sampling from live cells for temporal analysis. However, the side effects of these processes on gene expression, that can affect the physiology of sensitive stem cells are not well understood. Here, we use single cell RNA sequencing (scRNA-seq) to investigate the effects of BEP and LEP on murine neural stem cell (NSC) gene expression. Our results indicate that unlike BEP, LEP does not lead to extensive cell death or activation of cell stress response pathways that may affect their long-term physiology. Additionally, our demonstrations show that LEP is suitable for multi-day delivery protocols as it enables better preservation of cell viability and integrity as compared to BEP.
Collapse
|
26
|
Wan F, Dong Z, Liu B, Yan S, Wu N, Yang M, Chang L. Sensitive Interrogation of Enhancer Activity in Living Cells on a Nanoelectroporation-Probing Platform. ACS Sens 2022; 7:3671-3681. [PMID: 36410738 DOI: 10.1021/acssensors.2c01187] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Enhancers involved in the upregulation of multiple oncogenes play a fundamental role in tumorigenesis and immortalization. Exploring the activity of enhancers in living cells has emerged as a critical path to a deep understanding of cancer properties, further providing important clues to targeted therapy. However, identifying enhancer activity in living cells is challenging due to the double biological barriers of a cell cytoplasmic membrane and a nuclear membrane, limiting the sensitivity and responsiveness of conventional probing methods. In this work, we developed a nanoelectroporation-probing (NP) platform, which enables intranuclear probe delivery for sensitive interrogation of enhancer activity in living cells. The nanoelectroporation biochip achieved highly focused perforation of the cell cytoplasmic membrane and brought about additional driving force to expedite the delivery of probes into the nucleus. The probes targeting enhancer activity (named "PH probe") are programmed with a cyclic amplification strategy and enable an increase in the fluorescence signals over 100-fold within 1 h. The platform was leveraged to detect the activity of CCAT1 enhancers (CCAT1, colon cancer-associated transcript-1, a long noncoding RNA that functions in tumor invasion and metastasis) in cell samples from clinical lung cancer patients, as well as reveal the heterogeneity of enhancers among different patients. The observations may extend the linkages between enhancers and cancer cells while validating the robustness and reliability of the platform for the assay of enhancer activity. This platform will be a promising toolbox with wide applicable potential for the intranuclear study of living cells.
Collapse
Affiliation(s)
- Fengqi Wan
- Key Laboratory of Biomechanics and Mechanobiology (Ministry of Education), Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China.,Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou 730030, China
| | - Zaizai Dong
- Key Laboratory of Biomechanics and Mechanobiology (Ministry of Education), Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Bing Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Shi Yan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Nan Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Mingzhu Yang
- Key Laboratory of Biomechanics and Mechanobiology (Ministry of Education), Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Lingqian Chang
- Key Laboratory of Biomechanics and Mechanobiology (Ministry of Education), Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China.,School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
27
|
Mukherjee P, Park SH, Pathak N, Patino CA, Bao G, Espinosa HD. Integrating Micro and Nano Technologies for Cell Engineering and Analysis: Toward the Next Generation of Cell Therapy Workflows. ACS NANO 2022; 16:15653-15680. [PMID: 36154011 DOI: 10.1021/acsnano.2c05494] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The emerging field of cell therapy offers the potential to treat and even cure a diverse array of diseases for which existing interventions are inadequate. Recent advances in micro and nanotechnology have added a multitude of single cell analysis methods to our research repertoire. At the same time, techniques have been developed for the precise engineering and manipulation of cells. Together, these methods have aided the understanding of disease pathophysiology, helped formulate corrective interventions at the cellular level, and expanded the spectrum of available cell therapeutic options. This review discusses how micro and nanotechnology have catalyzed the development of cell sorting, cellular engineering, and single cell analysis technologies, which have become essential workflow components in developing cell-based therapeutics. The review focuses on the technologies adopted in research studies and explores the opportunities and challenges in combining the various elements of cell engineering and single cell analysis into the next generation of integrated and automated platforms that can accelerate preclinical studies and translational research.
Collapse
Affiliation(s)
- Prithvijit Mukherjee
- Department of Mechanical Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Theoretical and Applied Mechanics Program, Northwestern University, Evanston, Illinois 60208, United States
| | - So Hyun Park
- Department of Bioengineering, Rice University, 6500 Main Street, Houston, Texas 77030, United States
| | - Nibir Pathak
- Department of Mechanical Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Theoretical and Applied Mechanics Program, Northwestern University, Evanston, Illinois 60208, United States
| | - Cesar A Patino
- Department of Mechanical Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Gang Bao
- Department of Bioengineering, Rice University, 6500 Main Street, Houston, Texas 77030, United States
| | - Horacio D Espinosa
- Department of Mechanical Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Theoretical and Applied Mechanics Program, Northwestern University, Evanston, Illinois 60208, United States
| |
Collapse
|
28
|
Lard M, Ho BD, Beech JP, Tegenfeldt JO, Prinz CN. Use of dielectrophoresis for directing T cells to microwells before nanostraw transfection: modelling and experiments. RSC Adv 2022; 12:30295-30303. [PMID: 36337971 PMCID: PMC9589401 DOI: 10.1039/d2ra05119b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/14/2022] [Indexed: 11/07/2022] Open
Abstract
Nanostraw substrates have great potential for achieving minimally invasive cell transfection. Cells located on the nanostraw substrate are subjected to mild DC electric pulses applied across the nanostraw substrate, which open pores in the cell membrane on top of the nanostraws and drives charged cargo through these pores via electrophoresis. However, with this method, the current may leak through uncovered nanostraws, thereby decreasing the desired effect in the cell-covered nanostraws. A minimization of the number of uncovered nanostraws could be achieved by high cell coverage, but this is challenging when working with small cell populations. Nanostraw substrates of smaller area could be covered by smaller cell populations but are hard to integrate into fluidics systems. Here, we use simulations and experiments to show that this issue can be addressed by covering the nanostraw substrate with an insulating layer containing pores of similar size to cells. The pores act as traps into which cells can be guided using dielectrophoresis, ensuring a high degree of occupancy while maintaining a high cell viability, even if the total number of cells is low.
Collapse
Affiliation(s)
- Mercy Lard
- Division of Solid State Physics and NanoLund, Lund University 221 00 Lund Sweden
| | - Bao D Ho
- Division of Solid State Physics and NanoLund, Lund University 221 00 Lund Sweden
| | - Jason P Beech
- Division of Solid State Physics and NanoLund, Lund University 221 00 Lund Sweden
| | - Jonas O Tegenfeldt
- Division of Solid State Physics and NanoLund, Lund University 221 00 Lund Sweden
| | - Christelle N Prinz
- Division of Solid State Physics and NanoLund, Lund University 221 00 Lund Sweden
| |
Collapse
|
29
|
Hall RN, Weill U, Drees L, Leal-Ortiz S, Li H, Khariton M, Chai C, Xue Y, Rosental B, Quake SR, Sánchez Alvarado A, Melosh NA, Fire AZ, Rink JC, Wang B. Heterologous reporter expression in the planarian Schmidtea mediterranea through somatic mRNA transfection. CELL REPORTS METHODS 2022; 2:100298. [PMID: 36313809 PMCID: PMC9606109 DOI: 10.1016/j.crmeth.2022.100298] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 06/11/2022] [Accepted: 08/25/2022] [Indexed: 06/16/2023]
Abstract
Planarians have long been studied for their regenerative abilities. Moving forward, tools for ectopic expression of non-native proteins will be of substantial value. Using a luminescent reporter to overcome the strong autofluorescence of planarian tissues, we demonstrate heterologous protein expression in planarian cells and live animals. Our approach is based on the introduction of mRNA through several nanotechnological and chemical transfection methods. We improve reporter expression by altering untranslated region (UTR) sequences and codon bias, facilitating the measurement of expression kinetics in both isolated cells and whole planarians using luminescence imaging. We also examine protein expression as a function of variations in the UTRs of delivered mRNA, demonstrating a framework to investigate gene regulation at the post-transcriptional level. Together, these advances expand the toolbox for the mechanistic analysis of planarian biology and establish a foundation for the development and expansion of transgenic techniques in this unique model system.
Collapse
Affiliation(s)
| | - Uri Weill
- Department of Tissue Dynamics and Regeneration, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Leonard Drees
- Department of Tissue Dynamics and Regeneration, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Sergio Leal-Ortiz
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA
| | - Hongquan Li
- Department of Electrical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Margarita Khariton
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Chew Chai
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Yuan Xue
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Benyamin Rosental
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Center for Regenerative Medicine and Stem Cells, Ben Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Stephen R. Quake
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Department of Applied Physics, Stanford University, Stanford, CA 94305, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Alejandro Sánchez Alvarado
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
- Howard Hughes Medical Institute, Kansas City, MO 64110, USA
| | - Nicholas A. Melosh
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA
| | - Andrew Z. Fire
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jochen C. Rink
- Department of Tissue Dynamics and Regeneration, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Bo Wang
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
30
|
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
|
31
|
Elnathan R, Tay A, Voelcker NH, Chiappini C. The start-ups taking nanoneedles into the clinic. NATURE NANOTECHNOLOGY 2022; 17:807-811. [PMID: 35760894 DOI: 10.1038/s41565-022-01158-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Affiliation(s)
- Roey Elnathan
- School of Medicine, Faculty of Health, Deakin University, Waurn Ponds, Victoria, Australia.
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia.
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, Victoria, Australia.
| | - Andy Tay
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore
| | - Nicolas H Voelcker
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, Victoria, Australia
| | - Ciro Chiappini
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
- London Centre for Nanotechnology, King's College London, London, UK
| |
Collapse
|
32
|
Patino CA, Pathak N, Mukherjee P, Park SH, Bao G, Espinosa HD. Multiplexed high-throughput localized electroporation workflow with deep learning-based analysis for cell engineering. SCIENCE ADVANCES 2022; 8:eabn7637. [PMID: 35867793 PMCID: PMC9307252 DOI: 10.1126/sciadv.abn7637] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 06/07/2022] [Indexed: 05/06/2023]
Abstract
Manipulation of cells for applications such as biomanufacturing and cell-based therapeutics involves introducing biomolecular cargoes into cells. However, successful delivery is a function of multiple experimental factors requiring several rounds of optimization. Here, we present a high-throughput multiwell-format localized electroporation device (LEPD) assisted by deep learning image analysis that enables quick optimization of experimental factors for efficient delivery. We showcase the versatility of the LEPD platform by successfully delivering biomolecules into different types of adherent and suspension cells. We also demonstrate multicargo delivery with tight dosage distribution and precise ratiometric control. Furthermore, we used the platform to achieve functional gene knockdown in human induced pluripotent stem cells and used the deep learning framework to analyze protein expression along with changes in cell morphology. Overall, we present a workflow that enables combinatorial experiments and rapid analysis for the optimization of intracellular delivery protocols required for genetic manipulation.
Collapse
Affiliation(s)
- Cesar A. Patino
- Department of Mechanical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Nibir Pathak
- Department of Mechanical Engineering, Northwestern University, Evanston, IL 60208, USA
- Theoretical and Applied Mechanics Program, Northwestern University, Evanston, IL 60208, USA
| | - Prithvijit Mukherjee
- Department of Mechanical Engineering, Northwestern University, Evanston, IL 60208, USA
- Theoretical and Applied Mechanics Program, Northwestern University, Evanston, IL 60208, USA
| | - So Hyun Park
- Department of Bioengineering, Rice University, 6500 Main St, Houston, TX 77030, USA
| | - Gang Bao
- Department of Bioengineering, Rice University, 6500 Main St, Houston, TX 77030, USA
| | - Horacio D. Espinosa
- Department of Mechanical Engineering, Northwestern University, Evanston, IL 60208, USA
- Theoretical and Applied Mechanics Program, Northwestern University, Evanston, IL 60208, USA
| |
Collapse
|
33
|
Zhang A, Fang J, Wang J, Xie X, Chen HJ, He G. Interrogation on the Cellular Nano-Interface and Biosafety of Repeated Nano-Electroporation by Nanostraw System. BIOSENSORS 2022; 12:522. [PMID: 35884325 PMCID: PMC9313307 DOI: 10.3390/bios12070522] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/07/2022] [Accepted: 07/11/2022] [Indexed: 11/22/2022]
Abstract
Cell perforation is a critical step for intracellular drug delivery and real-time biosensing of intracellular signals. In recent years, the nanostraws system has been developed to achieve intracellular drug delivery with minimal invasiveness to the cells. Repeated cell perforation via nano-system could allow delivery of multiple drugs into cells for cell editing, but the biosafety is rarely explored. In this work, a nanostraw-mediated nano-electroporation system was developed, which allowed repeated perforation of the same set of cells in a minimally invasive manner, while the biosafety aspect of this system was investigated. Highly controllable fabrication of Al2O3 nanostraw arrays based on a porous polyethylene terephthalate (PET) membrane was integrated with a microfluidic device to construct the nanostraw-electroporation system. The pulse conditions and intervals of nano-electroporation were systematically optimized to achieve efficient cells perforation and maintain the viability of the cells. The cells proliferation, the early apoptosis activities after nanostraw-electroporation and the changes of gene functions and gene pathways of cells after repeated nano-electroporation were comprehensively analyzed. These results revealed that the repeated nanostraw-electroporation did not induce obvious negative effects on the cells. This work demonstrates the feasibility of repeated nano-electroporation on cells and provides a promising strategy for future biomedical applications.
Collapse
Affiliation(s)
- Aihua Zhang
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information, Technology, Sun Yat-sen University, Guangzhou 510006, China; (A.Z.); (J.F.); (X.X.)
| | - Jiaru Fang
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information, Technology, Sun Yat-sen University, Guangzhou 510006, China; (A.Z.); (J.F.); (X.X.)
| | - Ji Wang
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China;
| | - Xi Xie
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information, Technology, Sun Yat-sen University, Guangzhou 510006, China; (A.Z.); (J.F.); (X.X.)
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China;
| | - Hui-Jiuan Chen
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information, Technology, Sun Yat-sen University, Guangzhou 510006, China; (A.Z.); (J.F.); (X.X.)
| | - Gen He
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information, Technology, Sun Yat-sen University, Guangzhou 510006, China; (A.Z.); (J.F.); (X.X.)
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| |
Collapse
|
34
|
Esmek FM, Erichlandwehr T, Brkovic N, Pranzner NP, Teuber JP, Fernandez-Cuesta I. Pillar-structured 3D inlets fabricated by dose-modulated e-beam lithography and nanoimprinting for DNA analysis in passive, clogging-free, nanofluidic devices. NANOTECHNOLOGY 2022; 33:385301. [PMID: 35696945 DOI: 10.1088/1361-6528/ac780d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 06/13/2022] [Indexed: 06/15/2023]
Abstract
We present the fabrication of three-dimensional inlets with gradually decreasing widths and depths and with nanopillars on the slope, all defined in just one lithography step. In addition, as an application, we show how these micro- and nanostructures can be used for micro- and nanofluidics and lab-on-a-chip devices to facilitate the flow and analyze single molecules of DNA. For the fabrication of 3D inlets in a single layer process, dose-modulated electron beam lithography was used, producing depths between 750 nm and 50 nm along a 30 μm long inlet, which is additionally structured with nanometer-scale pillars randomly distributed on top, as a result of incomplete exposure and underdevelopment of the resist. The fabrication conditions affect the slope of the inlet, the nanopillar density and coverage. The key parameters are the dose used for the electron beam exposure and the development conditions, like the developer's dilution, stirring and development time. The 3D inlets with nanostructured pillars were integrated into fluidic devices, acting as a transition between micro and nanofluidic structures for pre-stretching and unfolding DNA molecules, avoiding the intrusion of folded molecules and clogging the analysis channel. After patterning these structures in silicon, they can be replicated in polymer by UV nanoimprinting. We show here how the inlets with pillars slow down the molecules before they enter the nanochannels, resulting in a 3-fold decrease in speed, which would translate to an improvement in the resolution for DNA optical mapping.
Collapse
Affiliation(s)
- Franziska M Esmek
- Universität Hamburg, Institute of Nanostructure and Solid State Physics, HARBOR Bldg 610, Luruper Chaussee 149, Hamburg D-22761, Germany
| | - Tim Erichlandwehr
- Universität Hamburg, Institute of Nanostructure and Solid State Physics, HARBOR Bldg 610, Luruper Chaussee 149, Hamburg D-22761, Germany
| | - Nico Brkovic
- Universität Hamburg, Institute of Nanostructure and Solid State Physics, HARBOR Bldg 610, Luruper Chaussee 149, Hamburg D-22761, Germany
| | - Nathalie P Pranzner
- Universität Hamburg, Institute of Nanostructure and Solid State Physics, HARBOR Bldg 610, Luruper Chaussee 149, Hamburg D-22761, Germany
| | - Jeremy P Teuber
- Universität Hamburg, Institute of Nanostructure and Solid State Physics, HARBOR Bldg 610, Luruper Chaussee 149, Hamburg D-22761, Germany
| | - Irene Fernandez-Cuesta
- Universität Hamburg, Institute of Nanostructure and Solid State Physics, HARBOR Bldg 610, Luruper Chaussee 149, Hamburg D-22761, Germany
- Hamburg Centre for Ultrafast Imaging, Germany
| |
Collapse
|
35
|
Jiang Z, Fu M, Zhu D, Wang X, Li N, Ren L, He J, Yang G. Genetically modified immunomodulatory cell-based biomaterials in tissue regeneration and engineering. Cytokine Growth Factor Rev 2022; 66:53-73. [PMID: 35690567 DOI: 10.1016/j.cytogfr.2022.05.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 05/24/2022] [Indexed: 11/25/2022]
Abstract
To date, the wide application of cell-based biomaterials in tissue engineering and regeneration is remarkably hampered by immune rejection. Reducing the immunogenicity of cell-based biomaterials has become the latest direction in biomaterial research. Recently, genetically modified cell-based biomaterials with immunomodulatory genes have become a feasible solution to the immunogenicity problem. In this review, recent advances and future challenges of genetically modified immunomodulatory cell-based biomaterials are elaborated, including fabrication approaches, mechanisms of common immunomodulatory genes, application and, more importantly, current preclinical and clinical advances. The fabrication approaches can be categorized into commonly used (e.g., virus transfection) and newly developed approaches. The immunomodulatory mechanisms of representative genes involve complicated cell signaling pathways and metabolic activities. Wide application in curing multiple end-term diseases and replacing lifelong immunosuppressive therapy in multiple cell and organ transplantation models is demonstrated. Most significantly, practices of genetically modified organ transplantation have been conducted on brain-dead human decedent and even on living patients after a series of experiments on nonhuman primates. Nevertheless, uncertain biosecurity, nonspecific effects and overlooked personalization of current genetically modified immunomodulatory cell-based biomaterials are shortcomings that remain to be overcome.
Collapse
Affiliation(s)
- Zhiwei Jiang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center of Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Mengdie Fu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center of Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Danji Zhu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center of Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Xueting Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center of Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Na Li
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center of Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Lingfei Ren
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center of Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Jin He
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center of Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Guoli Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center of Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China.
| |
Collapse
|
36
|
Chen N, He Y, Zang M, Zhang Y, Lu H, Zhao Q, Wang S, Gao Y. Approaches and materials for endocytosis-independent intracellular delivery of proteins. Biomaterials 2022; 286:121567. [DOI: 10.1016/j.biomaterials.2022.121567] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 04/26/2022] [Accepted: 05/03/2022] [Indexed: 12/12/2022]
|
37
|
Mukherjee P, Patino CA, Pathak N, Lemaitre V, Espinosa HD. Deep Learning-Assisted Automated Single Cell Electroporation Platform for Effective Genetic Manipulation of Hard-to-Transfect Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2107795. [PMID: 35315229 PMCID: PMC9119920 DOI: 10.1002/smll.202107795] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/03/2022] [Indexed: 05/03/2023]
Abstract
Genome engineering of cells using CRISPR/Cas systems has opened new avenues for pharmacological screening and investigating the molecular mechanisms of disease. A critical step in many such studies is the intracellular delivery of the gene editing machinery and the subsequent manipulation of cells. However, these workflows often involve processes such as bulk electroporation for intracellular delivery and fluorescence activated cell sorting for cell isolation that can be harsh to sensitive cell types such as human-induced pluripotent stem cells (hiPSCs). This often leads to poor viability and low overall efficacy, requiring the use of large starting samples. In this work, a fully automated version of the nanofountain probe electroporation (NFP-E) system, a nanopipette-based single-cell electroporation method is presented that provides superior cell viability and efficiency compared to traditional methods. The automated system utilizes a deep convolutional network to identify cell locations and a cell-nanopipette contact algorithm to position the nanopipette over each cell for the application of electroporation pulses. The automated NFP-E is combined with microconfinement arrays for cell isolation to demonstrate a workflow that can be used for CRISPR/Cas9 gene editing and cell tracking with potential applications in screening studies and isogenic cell line generation.
Collapse
Affiliation(s)
- Prithvijit Mukherjee
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Theoretical and Applied Mechanics Program, Northwestern University, Evanston, IL, 60208, USA
- iNfinitesimal LLC, Skokie, IL, 60077, USA
| | - Cesar A Patino
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, 60208, USA
- iNfinitesimal LLC, Skokie, IL, 60077, USA
| | - Nibir Pathak
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Theoretical and Applied Mechanics Program, Northwestern University, Evanston, IL, 60208, USA
| | | | - Horacio D Espinosa
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Theoretical and Applied Mechanics Program, Northwestern University, Evanston, IL, 60208, USA
- iNfinitesimal LLC, Skokie, IL, 60077, USA
| |
Collapse
|
38
|
Houthaeve G, De Smedt SC, Braeckmans K, De Vos WH. The cellular response to plasma membrane disruption for nanomaterial delivery. NANO CONVERGENCE 2022; 9:6. [PMID: 35103909 PMCID: PMC8807741 DOI: 10.1186/s40580-022-00298-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/05/2022] [Indexed: 06/14/2023]
Abstract
Delivery of nanomaterials into cells is of interest for fundamental cell biological research as well as for therapeutic and diagnostic purposes. One way of doing so is by physically disrupting the plasma membrane (PM). Several methods that exploit electrical, mechanical or optical cues have been conceived to temporarily disrupt the PM for intracellular delivery, with variable effects on cell viability. However, apart from acute cytotoxicity, subtler effects on cell physiology may occur as well. Their nature and timing vary with the severity of the insult and the efficiency of repair, but some may provoke permanent phenotypic alterations. With the growing palette of nanoscale delivery methods and applications, comes a need for an in-depth understanding of this cellular response. In this review, we summarize current knowledge about the chronology of cellular events that take place upon PM injury inflicted by different delivery methods. We also elaborate on their significance for cell homeostasis and cell fate. Based on the crucial nodes that govern cell fitness and functionality, we give directions for fine-tuning nano-delivery conditions.
Collapse
Affiliation(s)
- Gaëlle Houthaeve
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ghent, Belgium
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ghent, Belgium
| | - Kevin Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ghent, Belgium
| | - Winnok H De Vos
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
39
|
Transient nuclear lamin A/C accretion aids in recovery from vapor nanobubble-induced permeabilisation of the plasma membrane. Cell Mol Life Sci 2022; 79:23. [PMID: 34984553 PMCID: PMC8727414 DOI: 10.1007/s00018-021-04099-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 11/23/2021] [Accepted: 12/15/2021] [Indexed: 01/14/2023]
Abstract
Vapor nanobubble (VNB) photoporation is a physical method for intracellular delivery that has gained significant interest in the past decade. It has successfully been used to introduce molecular cargo of diverse nature into different cell types with high throughput and minimal cytotoxicity. For translational purposes, it is important to understand whether and how photoporation affects cell homeostasis. To obtain a comprehensive view on the transcriptional rewiring that takes place after VNB photoporation, we performed a longitudinal shotgun RNA-sequencing experiment. Six hours after photoporation, we found a marked upregulation of LMNA transcripts as well as their protein products, the A-type lamins. At the same time point, we observed a significant increase in several heterochromatin marks, suggesting a global stiffening of the nucleus. These molecular features vanished 24 h after photoporation. Since VNB-induced chromatin condensation was prolonged in LMNA knockout cells, A-type lamins may be required for restoring the nucleus to its original state. Selective depletion of A-type lamins reduced cell viability after VNB photoporation, while pharmacological stimulation of LMNA transcription increased the percentage of successfully transfected cells that survived after photoporation. Therefore, our results suggest that cells respond to VNB photoporation by temporary upregulation of A-type lamins to facilitate their recovery.
Collapse
|
40
|
Chakrabarty P, Gupta P, Illath K, Kar S, Nagai M, Tseng FG, Santra TS. Microfluidic mechanoporation for cellular delivery and analysis. Mater Today Bio 2022; 13:100193. [PMID: 35005598 PMCID: PMC8718663 DOI: 10.1016/j.mtbio.2021.100193] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/13/2021] [Accepted: 12/20/2021] [Indexed: 01/08/2023] Open
Abstract
Highly efficient intracellular delivery strategies are essential for developing therapeutic, diagnostic, biological, and various biomedical applications. The recent advancement of micro/nanotechnology has focused numerous researches towards developing microfluidic device-based strategies due to the associated high throughput delivery, cost-effectiveness, robustness, and biocompatible nature. The delivery strategies can be carrier-mediated or membrane disruption-based, where membrane disruption methods find popularity due to reduced toxicity, enhanced delivery efficiency, and cell viability. Among all of the membrane disruption techniques, the mechanoporation strategies are advantageous because of no external energy source required for membrane deformation, thereby achieving high delivery efficiencies and increased cell viability into different cell types with negligible toxicity. The past two decades have consequently seen a tremendous boost in mechanoporation-based research for intracellular delivery and cellular analysis. This article provides a brief review of the most recent developments on microfluidic-based mechanoporation strategies such as microinjection, nanoneedle arrays, cell-squeezing, and hydroporation techniques with their working principle, device fabrication, cellular delivery, and analysis. Moreover, a brief discussion of the different mechanoporation strategies integrated with other delivery methods has also been provided. Finally, the advantages, limitations, and future prospects of this technique are discussed compared to other intracellular delivery techniques.
Collapse
Affiliation(s)
- Pulasta Chakrabarty
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai, India
| | - Pallavi Gupta
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai, India
| | - Kavitha Illath
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai, India
| | - Srabani Kar
- Department of Electrical Engineering, University of Cambridge, Cambridge, CB30FA, UK
| | - Moeto Nagai
- Department of Mechanical Engineering, Toyohashi University of Technology, Aichi, Japan
| | - Fan-Gang Tseng
- Department of Engineering and System Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Tuhin Subhra Santra
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai, India
| |
Collapse
|
41
|
Harberts J, Bours K, Siegmund M, Hedrich C, Glatza M, Schöler HR, Haferkamp U, Pless O, Zierold R, Blick RH. Culturing human iPSC-derived neural progenitor cells on nanowire arrays: mapping the impact of nanowire length and array pitch on proliferation, viability, and membrane deformation. NANOSCALE 2021; 13:20052-20066. [PMID: 34842880 DOI: 10.1039/d1nr04352h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Nanowire arrays used as cell culture substrates build a potent tool for advanced biological applications such as cargo delivery and biosensing. The unique topography of nanowire arrays, however, renders them a challenging growth environment for cells and explains why only basic cell lines have been employed in existing studies. Here, we present the culturing of human induced pluripotent stem cell-derived neural progenitor cells on rectangularly arranged nanowire arrays: In detail, we mapped the impact on proliferation, viability, and topography-induced membrane deformation across a multitude of array pitches (1, 3, 5, 10 μm) and nanowire lengths (1.5, 3, 5 μm). Against the intuitive expectation, a reduced proliferation was found on the arrays with the smallest array pitch of 1 μm and long NWs. Typically, cells settle in a fakir-like state on such densely-spaced nanowires and thus experience no substantial stress caused by nanowires indenting the cell membrane. However, imaging of F-actin showed a distinct reorganization of the cytoskeleton along the nanowire tips in the case of small array pitches interfering with regular proliferation. For larger pitches, the cell numbers depend on the NW lengths but proliferation generally continued although heavy deformations of the cell membrane were observed caused by the encapsulation of the nanowires. Moreover, we noticed a strong interaction of the nanowires with the nucleus in terms of squeezing and indenting. Remarkably, the cell viability is maintained at about 85% despite the massive deformation of the cells. Considering the enormous potential of human induced stem cells to study neurodegenerative diseases and the high cellular viability combined with a strong interaction with nanowire arrays, we believe that our results pave the way to apply nanowire arrays to human stem cells for future applications in stem cell research and regenerative medicine.
Collapse
Affiliation(s)
- Jann Harberts
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany.
| | - Katja Bours
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany.
| | - Malte Siegmund
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany.
| | - Carina Hedrich
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany.
| | - Michael Glatza
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Hans R Schöler
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Undine Haferkamp
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), ScreeningPort, Schnackenburgallee 114, 22525 Hamburg, Germany
| | - Ole Pless
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), ScreeningPort, Schnackenburgallee 114, 22525 Hamburg, Germany
| | - Robert Zierold
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany.
| | - Robert H Blick
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany.
- Material Science and Engineering, College of Engineering, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| |
Collapse
|
42
|
Dong Z, Chang L. Recent electroporation-based systems for intracellular molecule delivery. NANOTECHNOLOGY AND PRECISION ENGINEERING 2021. [DOI: 10.1063/10.0005649] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Zaizai Dong
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Lingqian Chang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| |
Collapse
|
43
|
Microfluidic transfection of mRNA into human primary lymphocytes and hematopoietic stem and progenitor cells using ultra-fast physical deformations. Sci Rep 2021; 11:21407. [PMID: 34725429 PMCID: PMC8560772 DOI: 10.1038/s41598-021-00893-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 10/19/2021] [Indexed: 01/22/2023] Open
Abstract
Messenger RNA (mRNA) delivery provides gene therapy with the potential to achieve transient therapeutic efficacy without risk of insertional mutagenesis. Amongst other applications, mRNA can be employed as a platform to deliver gene editing molecules, to achieve protein expression as an alternative to enzyme replacement therapies, and to express chimeric antigen receptors (CARs) on immune cells for the treatment of cancer. We designed a novel microfluidic device that allows for efficient mRNA delivery via volume exchange for convective transfection (VECT). In the device, cells flow through a ridged channel that enforces a series of ultra-fast and large intensity deformations able to transiently open pores and induce convective transport of mRNA into the cell. Here, we describe efficient delivery of mRNA into T cells, natural killer (NK) cells and hematopoietic stem and progenitor cells (HSPCs), three human primary cell types widely used for ex vivo gene therapy applications. Results demonstrate that the device can operate at a wide range of cell and payload concentrations and that ultra-fast compressions do not have a negative impact on T cell function, making this a novel and competitive platform for the development of ex vivo mRNA-based gene therapies and other cell products engineered with mRNA.
Collapse
|
44
|
Chiappini C, Chen Y, Aslanoglou S, Mariano A, Mollo V, Mu H, De Rosa E, He G, Tasciotti E, Xie X, Santoro F, Zhao W, Voelcker NH, Elnathan R. Tutorial: using nanoneedles for intracellular delivery. Nat Protoc 2021; 16:4539-4563. [PMID: 34426708 DOI: 10.1038/s41596-021-00600-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 06/30/2021] [Indexed: 02/08/2023]
Abstract
Intracellular delivery of advanced therapeutics, including biologicals and supramolecular agents, is complex because of the natural biological barriers that have evolved to protect the cell. Efficient delivery of therapeutic nucleic acids, proteins, peptides and nanoparticles is crucial for clinical adoption of emerging technologies that can benefit disease treatment through gene and cell therapy. Nanoneedles are arrays of vertical high-aspect-ratio nanostructures that can precisely manipulate complex processes at the cell interface, enabling effective intracellular delivery. This emerging technology has already enabled the development of efficient and non-destructive routes for direct access to intracellular environments and delivery of cell-impermeant payloads. However, successful implementation of this technology requires knowledge of several scientific fields, making it complex to access and adopt by researchers who are not directly involved in developing nanoneedle platforms. This presents an obstacle to the widespread adoption of nanoneedle technologies for drug delivery. This tutorial aims to equip researchers with the knowledge required to develop a nanoinjection workflow. It discusses the selection of nanoneedle devices, approaches for cargo loading and strategies for interfacing to biological systems and summarises an array of bioassays that can be used to evaluate the efficacy of intracellular delivery.
Collapse
Affiliation(s)
- Ciro Chiappini
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK.
- London Centre for Nanotechnology, King's College London, London, UK.
| | - Yaping Chen
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, Victoria, Australia
| | - Stella Aslanoglou
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, Victoria, Australia
- CSIRO Manufacturing, Clayton, Victoria, Australia
| | - Anna Mariano
- Center for Advanced Biomaterials for Healthcare, Istituto Italiano di Tecnologia, Naples, Italy
| | - Valentina Mollo
- Center for Advanced Biomaterials for Healthcare, Istituto Italiano di Tecnologia, Naples, Italy
| | - Huanwen Mu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| | - Enrica De Rosa
- Center for Musculoskeletal Regeneration, Orthopedics & Sports Medicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Gen He
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou, China
| | - Ennio Tasciotti
- IRCCS San Raffaele Pisana Hospital, Rome, Italy
- San Raffaele University, Rome, Italy
- Sclavo Pharma, Siena, Italy
| | - Xi Xie
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou, China.
| | - Francesca Santoro
- Center for Advanced Biomaterials for Healthcare, Istituto Italiano di Tecnologia, Naples, Italy.
| | - Wenting Zhao
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore.
| | - Nicolas H Voelcker
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia.
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, Victoria, Australia.
- CSIRO Manufacturing, Clayton, Victoria, Australia.
- Department of Materials Science and Engineering, Monash University, Clayton, Victoria, Australia.
| | - Roey Elnathan
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia.
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, Victoria, Australia.
- Department of Materials Science and Engineering, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
45
|
Hur J, Chung AJ. Microfluidic and Nanofluidic Intracellular Delivery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2004595. [PMID: 34096197 PMCID: PMC8336510 DOI: 10.1002/advs.202004595] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 04/14/2021] [Indexed: 05/05/2023]
Abstract
Innate cell function can be artificially engineered and reprogrammed by introducing biomolecules, such as DNAs, RNAs, plasmid DNAs, proteins, or nanomaterials, into the cytosol or nucleus. This process of delivering exogenous cargos into living cells is referred to as intracellular delivery. For instance, clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 gene editing begins with internalizing Cas9 protein and guide RNA into cells, and chimeric antigen receptor-T (CAR-T) cells are prepared by delivering CAR genes into T lymphocytes for cancer immunotherapies. To deliver external biomolecules into cells, tools, including viral vectors, and electroporation have been traditionally used; however, they are suboptimal for achieving high levels of intracellular delivery while preserving cell viability, phenotype, and function. Notably, as emerging solutions, microfluidic and nanofluidic approaches have shown remarkable potential for addressing this open challenge. This review provides an overview of recent advances in microfluidic and nanofluidic intracellular delivery strategies and discusses new opportunities and challenges for clinical applications. Furthermore, key considerations for future efforts to develop microfluidics- and nanofluidics-enabled next-generation intracellular delivery platforms are outlined.
Collapse
Affiliation(s)
- Jeongsoo Hur
- School of Biomedical EngineeringKorea UniversitySeoul02841Republic of Korea
| | - Aram J. Chung
- School of Biomedical EngineeringInterdisciplinary Program in Precision Public HealthKorea UniversitySeoul02841Republic of Korea
| |
Collapse
|
46
|
Dong Z, Yan S, Liu B, Hao Y, Lin L, Chang T, Sun H, Wang Y, Li H, Wu H, Hang X, He S, Hu J, Xue X, Wu N, Chang L. Single Living Cell Analysis Nanoplatform for High-Throughput Interrogation of Gene Mutation and Cellular Behavior. NANO LETTERS 2021; 21:4878-4886. [PMID: 33830766 DOI: 10.1021/acs.nanolett.1c00199] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The genetic heterogeneities in cancer cells pose challenges to achieving precise drug treatment in a widely applicable manner. Most single-cell gene analysis methods rely on cell lysis for gene extraction and identification, showing limited capacity to provide the correlation of genetic properties and real-time cellular behaviors. Here, we report a single living cell analysis nanoplatform that enables interrogating gene properties and drug resistance in millions of single cells. We designed a Domino-probe to identify intracellular target RNAs while releasing 10-fold amplified fluorescence signals. An on-chip addressable microwell-nanopore array was developed for enhanced electro-delivery of the Domino-probe and in situ observation of cell behaviors. The proof-of-concept of the system was validated in primary lung cancer cell samples, revealing the positive-correlation of the ratio of EGFR mutant cells with their drug susceptibilities. This platform provides a high-throughput yet precise tool for exploring the relationship between intracellular genes and cell behaviors at the single-cell level.
Collapse
Affiliation(s)
- Zaizai Dong
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Shi Yan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Bing Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yongcun Hao
- Ministry of Education Key Laboratory of Micro and Nano Systems for Aerospace, Northwestern Polytechnical University, Xi'an 710072, China
| | - Long Lin
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Tianrui Chang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Hong Sun
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Yusen Wang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Hu Li
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Han Wu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Xinxin Hang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Shiqi He
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Jiaming Hu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Xinying Xue
- Department of Respiratory and Critical Care, Beijing Shijitan Hospital, Capital Medical University, Peking University Ninth School of Clinical Medicine, Beijing 100038, China
- Department of Respiratory and Critical Care, Chinese PLA General Hospital, Beijing 100853, China
- Affiliated Hospital of Weifang Medical University, Shandong 261031, China
| | - Nan Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Lingqian Chang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| |
Collapse
|
47
|
Raes L, De Smedt SC, Raemdonck K, Braeckmans K. Non-viral transfection technologies for next-generation therapeutic T cell engineering. Biotechnol Adv 2021; 49:107760. [PMID: 33932532 DOI: 10.1016/j.biotechadv.2021.107760] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 04/24/2021] [Accepted: 04/24/2021] [Indexed: 12/24/2022]
Abstract
Genetically engineered T cells have sparked interest in advanced cancer treatment, reaching a milestone in 2017 with two FDA-approvals for CD19-directed chimeric antigen receptor (CAR) T cell therapeutics. It is becoming clear that the next generation of CAR T cell therapies will demand more complex engineering strategies and combinations thereof, including the use of revolutionary gene editing approaches. To date, manufacturing of CAR T cells mostly relies on γ-retroviral or lentiviral vectors, but their use is associated with several drawbacks, including safety issues, high manufacturing cost and vector capacity constraints. Non-viral approaches, including membrane permeabilization and carrier-based techniques, have therefore gained a lot of interest to replace viral transductions in the manufacturing of T cell therapeutics. This review provides an in-depth discussion on the avid search for alternatives to viral vectors, discusses key considerations for T cell engineering technologies, and provides an overview of the emerging spectrum of non-viral transfection technologies for T cells. Strengths and weaknesses of each technology will be discussed in relation to T cell engineering. Altogether, this work emphasizes the potential of non-viral transfection approaches to advance the next-generation of genetically engineered T cells.
Collapse
Affiliation(s)
- Laurens Raes
- Laboratory of General Biochemistry & Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry & Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Koen Raemdonck
- Laboratory of General Biochemistry & Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Kevin Braeckmans
- Laboratory of General Biochemistry & Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| |
Collapse
|
48
|
Hebisch E, Hjort M, Volpati D, Prinz CN. Nanostraw-Assisted Cellular Injection of Fluorescent Nanodiamonds via Direct Membrane Opening. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2006421. [PMID: 33502091 DOI: 10.1002/smll.202006421] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/16/2020] [Indexed: 06/12/2023]
Abstract
Due to their stable fluorescence, biocompatibility, and amenability to functionalization, fluorescent nanodiamonds (FND) are promising materials for long term cell labeling and tracking. However, transporting them to the cytosol remains a major challenge, due to low internalization efficiencies and endosomal entrapment. Here, nanostraws in combination with low voltage electroporation pulses are used to achieve direct delivery of FND to the cytosol. The nanostraw delivery leads to efficient and rapid FND transport into cells compared to when incubating cells in a FND-containing medium. Moreover, whereas all internalized FND delivered by incubation end up in lysosomes, a significantly larger proportion of nanostraw-injected FND are in the cytosol, which opens up for using FND as cellular probes. Furthermore, in order to answer the long-standing question in the field of nano-biology regarding the state of the cell membrane on hollow nanostructures, live cell stimulated emission depletion (STED) microscopy is performed to image directly the state of the membrane on nanostraws. The time-lapse STED images reveal that the cell membrane opens entirely on top of nanostraws upon application of gentle electrical pulses, which supports the hypothesis that many FND are delivered directly to the cytosol, avoiding endocytosis and lysosomal entrapment.
Collapse
Affiliation(s)
- Elke Hebisch
- Division of Solid State Physics and NanoLund, Lund University, Lund, 221 00, Sweden
| | - Martin Hjort
- Division of Solid State Physics and NanoLund, Lund University, Lund, 221 00, Sweden
- Navan Technologies Inc., 733 Industrial Rd, San Carlos, CA, United States
| | - Diogo Volpati
- Division of Solid State Physics and NanoLund, Lund University, Lund, 221 00, Sweden
| | - Christelle N Prinz
- Division of Solid State Physics and NanoLund, Lund University, Lund, 221 00, Sweden
| |
Collapse
|