1
|
Mariello M, Eş I, Proctor CM. Soft and Flexible Bioelectronic Micro-Systems for Electronically Controlled Drug Delivery. Adv Healthc Mater 2024; 13:e2302969. [PMID: 37924224 DOI: 10.1002/adhm.202302969] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/20/2023] [Indexed: 11/06/2023]
Abstract
The concept of targeted and controlled drug delivery, which directs treatment to precise anatomical sites, offers benefits such as fewer side effects, reduced toxicity, optimized dosages, and quicker responses. However, challenges remain to engineer dependable systems and materials that can modulate host tissue interactions and overcome biological barriers. To stay aligned with advancements in healthcare and precision medicine, novel approaches and materials are imperative to improve effectiveness, biocompatibility, and tissue compliance. Electronically controlled drug delivery (ECDD) has recently emerged as a promising approach to calibrated drug delivery with spatial and temporal precision. This article covers recent breakthroughs in soft, flexible, and adaptable bioelectronic micro-systems designed for ECDD. It overviews the most widely reported operational modes, materials engineering strategies, electronic interfaces, and characterization techniques associated with ECDD systems. Further, it delves into the pivotal applications of ECDD in wearable, ingestible, and implantable medical devices. Finally, the discourse extends to future prospects and challenges for ECDD.
Collapse
Affiliation(s)
- Massimo Mariello
- Department of Engineering Science, Institute of Biomedical Engineering (IBME), University of Oxford, Oxford, OX3 7DQ, UK
| | - Ismail Eş
- Department of Engineering Science, Institute of Biomedical Engineering (IBME), University of Oxford, Oxford, OX3 7DQ, UK
| | - Christopher M Proctor
- Department of Engineering Science, Institute of Biomedical Engineering (IBME), University of Oxford, Oxford, OX3 7DQ, UK
| |
Collapse
|
2
|
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
ConspectusIn 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
|
3
|
Dolai J, Sarkar AR, Maity A, Mukherjee B, Jana NR. Ultrasonic Electroporation for Cells Grown on Piezoelectric Film Composed of Hydroxyapatite Nanowire and PVDF. ACS APPLIED MATERIALS & INTERFACES 2023; 15:59155-59164. [PMID: 38100427 DOI: 10.1021/acsami.3c13005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2023]
Abstract
The delivery of cell impermeable exogenous material into live cells by external stimuli is critical for both biological research and therapeutic applications. Although electroporation-based delivery of foreign materials inside the cell is a powerful approach, cell viability is often compromised due to the requirement of high voltage. Here, we report a piezoelectric hydroxyapatite nanowire-embedded poly(vinylidene fluoride) (PVDF) film for ultrasonic electroporation-based delivery of foreign materials to adherent cells. We found that 9 wt % loading of hydroxyapatite nanowires into PVDF can enhance the piezoelectric property by 2-3 times (with a piezoelectric constant value of 58 pm/V) than pure PVDF/nanowire, which is comparable to commonly known piezoelectric ceramics. These films can harvest mechanical as well as ultrasound-based energy to produce electrical potential up to 2 V. This biocompatible film can be used to grow cells on top of it and for subsequent application of ultrasound to exert electric voltage on cell membrane. We found that ultrasonic exposure to adhered cells leads to reversible pore formation on cell membrane that offers intracellular delivery of FITC-dextran with 75% efficiency. The developed piezoelectric film-based ultrasonic electroporation can be used for wireless electroporation in remote areas.
Collapse
Affiliation(s)
- Jayanta Dolai
- School of Materials Science, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Kolkata 700032, India
| | - Abu Raihan Sarkar
- School of Materials Science, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Kolkata 700032, India
| | - Anupam Maity
- School of Materials Science, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Kolkata 700032, India
| | - Buddhadev Mukherjee
- School of Materials Science, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Kolkata 700032, India
| | - Nikhil R Jana
- School of Materials Science, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Kolkata 700032, India
| |
Collapse
|
4
|
Park W, Kim EM, Jeon Y, Lee J, Yi J, Jeong J, Kim B, Jeong BG, Kim DR, Kong H, Lee CH. Transparent Intracellular Sensing Platform with Si Needles for Simultaneous Live Imaging. ACS NANO 2023; 17:25014-25026. [PMID: 38059775 DOI: 10.1021/acsnano.3c07527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/08/2023]
Abstract
Vertically ordered Si needles are of particular interest for long-term intracellular recording owing to their capacity to infiltrate living cells with negligible damage and minimal toxicity. Such intracellular recordings could greatly benefit from simultaneous live cell imaging without disrupting their culture, contributing to an in-depth understanding of cellular function and activity. However, the use of standard live imaging techniques, such as inverted and confocal microscopy, is currently impeded by the opacity of Si wafers, typically employed for fabricating vertical Si needles. Here, we introduce a transparent intracellular sensing platform that combines vertical Si needles with a percolated network of Au-Ag nanowires on a transparent elastomeric substrate. This sensing platform meets all prerequisites for simultaneous intracellular recording and imaging, including electrochemical impedance, optical transparency, mechanical compliance, and cell viability. Proof-of-concept demonstrations of this sensing platform include monitoring electrical potentials in cardiomyocyte cells and in three-dimensionally engineered cardiovascular tissue, all while conducting live imaging with inverted and confocal microscopes. This sensing platform holds wide-ranging potential applications for intracellular research across various disciplines such as neuroscience, cardiology, muscle physiology, and drug screening.
Collapse
Affiliation(s)
- Woohyun Park
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Eun Mi Kim
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Yale Jeon
- School of Mechanical Engineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Junsang Lee
- School of Mechanical Engineering, Hanyang University, Seoul 04763, Republic of Korea
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Jonghun Yi
- School of Mechanical Engineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Jinheon Jeong
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Bongjoong Kim
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
- Department of Mechanical and System Design Engineering, Hongik University, Seoul 04066, Republic of Korea
| | - Byeong Guk Jeong
- School of Mechanical Engineering, Hanyang University, Seoul 04763, Republic of Korea
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Dong Rip Kim
- School of Mechanical Engineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Hyunjoon Kong
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Chi Hwan Lee
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
- Department of Materials Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
5
|
Tang H, Yang H, Zhu W, Fei L, Huang J, Liu Z, Wang L, Chen H. Universal Strategy of Efficient Intracellular Macromolecule Directional Delivery Using Photothermal Pump Patch. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2304365. [PMID: 37594731 DOI: 10.1002/adma.202304365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/11/2023] [Indexed: 08/19/2023]
Abstract
The development of a highly efficient, nondestructive, and in vitro/vivo-applicable universal delivery strategy of therapeutic macromolecules into desired cells and tissues is very challenging. Photothermal methods have advantages in intracellular delivery, particularly in in vivo manipulation. However, the inability of directional transmission of exogenous molecules limits their delivery efficiency. Here, a photothermal pump (PTP) patch with numerous "exogenous molecular reservoirs" is reported. Under a laser, the cell membrane ruptures, while "exogenous molecular reservoirs" shrink, resulting in a directional exogenous molecule delivery into cells for a high-efficient intracellular delivery. The PTP patches are considered a universal structure for a highly efficient, nondestructive, and in-vitro/vivo-applicable intracellular macromolecule delivery. Under in vivo transdermal intracellular delivery conditions, the target genes are efficiently and noninvasively delivered into epidermal and dermal cells through the PTP patch and exosomes produced by the epidermal cells, respectively. The PTP patch provides a new strategy for a high-efficiency, nondestructive, and in-vitro/vivo-applicable macromolecule delivery.
Collapse
Affiliation(s)
- Heming Tang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - He Yang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
| | - Wenjun Zhu
- Institute of Functional Nano & Soft Materials Laboratory (FUNSOM), Soochow University, Suzhou, Jiangsu, 215123, China
| | - Liyan Fei
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Jialei Huang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials Laboratory (FUNSOM), Soochow University, Suzhou, Jiangsu, 215123, China
| | - Lei Wang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
- Jiangsu Biosurf Biotech Company Ltd., Suzhou, Jiangsu, 215123, China
| | - Hong Chen
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
| |
Collapse
|
6
|
Lv S, He E, Luo J, Liu Y, Liang W, Xu S, Zhang K, Yang Y, Wang M, Song Y, Wu Y, Cai X. Using Human-Induced Pluripotent Stem Cell Derived Neurons on Microelectrode Arrays to Model Neurological Disease: A Review. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301828. [PMID: 37863819 PMCID: PMC10667858 DOI: 10.1002/advs.202301828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 09/04/2023] [Indexed: 10/22/2023]
Abstract
In situ physiological signals of in vitro neural disease models are essential for studying pathogenesis and drug screening. Currently, an increasing number of in vitro neural disease models are established using human-induced pluripotent stem cell (hiPSC) derived neurons (hiPSC-DNs) to overcome interspecific gene expression differences. Microelectrode arrays (MEAs) can be readily interfaced with two-dimensional (2D), and more recently, three-dimensional (3D) neural stem cell-derived in vitro models of the human brain to monitor their physiological activity in real time. Therefore, MEAs are emerging and useful tools to model neurological disorders and disease in vitro using human iPSCs. This is enabling a real-time window into neuronal signaling at the network scale from patient derived. This paper provides a comprehensive review of MEA's role in analyzing neural disease models established by hiPSC-DNs. It covers the significance of MEA fabrication, surface structure and modification schemes for hiPSC-DNs culturing and signal detection. Additionally, this review discusses advances in the development and use of MEA technology to study in vitro neural disease models, including epilepsy, autism spectrum developmental disorder (ASD), and others established using hiPSC-DNs. The paper also highlights the application of MEAs combined with hiPSC-DNs in detecting in vitro neurotoxic substances. Finally, the future development and outlook of multifunctional and integrated devices for in vitro medical diagnostics and treatment are discussed.
Collapse
Affiliation(s)
- Shiya Lv
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Enhui He
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
- The State Key Lab of Brain‐Machine IntelligenceZhejiang UniversityHangzhou321100China
| | - Jinping Luo
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yaoyao Liu
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Wei Liang
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Shihong Xu
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Kui Zhang
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yan Yang
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Mixia Wang
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yilin Song
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yirong Wu
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Xinxia Cai
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| |
Collapse
|
7
|
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: 2] [Impact Index Per Article: 2.0] [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
|
8
|
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: 9] [Impact Index Per Article: 9.0] [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
|
9
|
Feng W, Wang Z, Tsubokawa M. Evaluations of nanoparticle capture and transport methods in dielectric bowtie core capillaries. OPTICS EXPRESS 2023; 31:16676-16689. [PMID: 37157742 DOI: 10.1364/oe.489355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
We propose a flexibly tunable and low-loss optical burette with an all-dielectric bowtie core capillary structure, where nanoparticle arrays can be transported bidirectionally with incident light from one end. Multiple hot spots, acting as optical traps, are periodically distributed at the center of the bowtie cores along the propagation direction because of the mode interference effect of guided lights. By adjusting the beam waist position, the hot spots continuously move across the entire capillary length; thus, trapped nanoparticles also transfer with the hot spots. The bidirectional transfer can be realized simply by changing the beam waist in the forward or backward direction. We confirmed that nanosized polystyrene spheres can be bidirectionally moved along a capillary length of ≈ 20 µm. Furthermore, the magnitude of the optical force can be adjusted using the incident angle and beam waist width, whereas the trapping period can be adjusted using the incident wavelength. These results were evaluated using the finite-difference time-domain method. We believe that this new approach can be extensively used in the field of biochemical and life sciences because of the properties of an all-dielectric structure, bidirectional transportation, and single incident light.
Collapse
|
10
|
Shinde A, Shinde P, Kar S, Illath K, Dey S, Mahapatra NR, Nagai M, Santra TS. Metallic micro-ring device for highly efficient large cargo delivery in mammalian cells using infrared light pulses. LAB ON A CHIP 2023; 23:2175-2192. [PMID: 36928187 DOI: 10.1039/d2lc00899h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Uniform transfection of biomolecules into live cells with high delivery efficiency and cell viability is an immensely important area of biological research and has many biomedical applications. In the present study, we report highly efficient, uniform parallel intracellular delivery of small to very large biomolecules into diverse cell types using a titanium micro-ring (TMR) device activated by infrared (IR) light pulse. A TMR array device (2 cm × 2 cm) consists of a 10 μm outer diameter and 3 μm inner diameter for each micro-ring, and 10 μm interspacing between two micro-rings. Upon IR (1050 nm) pulse laser irradiation on the TMR device, photothermal cavitation bubbles are generated, disrupting the cell plasma membrane, and biomolecules are gently delivered into the cells by a simple diffusion process. This TMR device successfully delivered diverse types of small to very large biomolecules such as propidium iodide (PI; 668.4 Da) dye, dextran (3 kDa), small interfering RNA (13.3 kDa), enhanced green fluorescent protein expression plasmid DNA (6.2 kb), and β-galactosidase enzyme (465 kDa) into human cervical (SiHa), mouse fibroblast (L929), and mouse neural crest-derived (N2a) cancer cells. For smaller molecules (PI dye), delivery efficiency and cell viability were achieved at ∼96% and ∼97%, respectively, with a laser fluence of 21 mJ cm-2 for 250 pulses. In contrast, ∼85% transfection efficiency and ∼90% cell viability were achieved for plasmid DNA with 45 mJ cm-2 laser fluence for 250 pulses in SiHa cells. Moreover, the intracellular delivery of β-galactosidase enzyme was confirmed with confocal microscopy and flow cytometry analysis resulting in ∼83% co-staining of β-galactosidase enzyme and calcein AM. Based on these efficient deliveries of diverse types of biomolecules in different cell types, the device has the potential for cellular diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Ashwini Shinde
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai, India.
| | - Pallavi Shinde
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai, India.
| | - Srabani Kar
- Indian Institute of Science Education and Research, Tirupati, India
| | - Kavitha Illath
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai, India.
| | - Souvik Dey
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai, India
| | - Nitish R Mahapatra
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai, India
| | - Moeto Nagai
- Department of Mechanical Engineering, Toyohashi University of Technology, Japan
| | - Tuhin Subhra Santra
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai, India.
| |
Collapse
|
11
|
Chakrabarty P, Illath K, Kar S, Nagai M, Santra TS. Combinatorial physical methods for cellular therapy: Towards the future of cellular analysis? J Control Release 2023; 353:1084-1095. [PMID: 36538949 DOI: 10.1016/j.jconrel.2022.12.038] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 12/16/2022] [Indexed: 12/25/2022]
Abstract
The physical energy activated techniques for cellular delivery and analysis is one of the most rapidly expanding research areas for a variety of biological and biomedical discoveries. These methods, such as electroporation, optoporation, sonoporation, mechanoporation, magnetoporation, etc., have been widely used in delivering different biomolecules into a range of primary and patient-derived cell types. However, the techniques when used individually have had limitations in delivery and co-delivery of diverse biomolecules in various cell types. In recent years, a number of studies have been performed by combining the different membrane disruption techniques, either sequentially or simultaneously, in a single study. The studies, referred to as combinatorial, or hybrid techniques, have demonstrated enhanced transfection, such as efficient macromolecular and gene delivery and co-delivery, at lower delivery parameters and with high cell viability. Such studies can open up new and exciting avenues for understanding the subcellular structure and consequently facilitate the development of novel therapeutic strategies. This review consequently aims at summarising the different developments in hybrid therapeutic techniques. The different methods discussed include mechano-electroporation, electro-sonoporation, magneto-mechanoporation, magnetic nanoparticles enhanced electroporation, and magnetic hyperthermia studies. We discuss the clinical status of the different methods and conclude with a discussion on the future prospects of the combinatorial techniques for cellular therapy and diagnostics.
Collapse
Affiliation(s)
- Pulasta Chakrabarty
- 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 Physics, Indian Institute of Science Education and Research, Tirupati, India
| | - Moeto Nagai
- Department of Mechanical Engineering, Toyohashi University of Technology, Aichi, Japan
| | - Tuhin Subhra Santra
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai, India.
| |
Collapse
|
12
|
Liu Y, Fan Z, Qiao L, Liu B. Advances in microfluidic strategies for single-cell research. Trends Analyt Chem 2022. [DOI: 10.1016/j.trac.2022.116822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
13
|
Abstract
Electroporation (EP) is a commonly used strategy to increase cell permeability for intracellular cargo delivery or irreversible cell membrane disruption using electric fields. In recent years, EP performance has been improved by shrinking electrodes and device structures to the microscale. Integration with microfluidics has led to the design of devices performing static EP, where cells are fixed in a defined region, or continuous EP, where cells constantly pass through the device. Each device type performs superior to conventional, macroscale EP devices while providing additional advantages in precision manipulation (static EP) and increased throughput (continuous EP). Microscale EP is gentle on cells and has enabled more sensitive assaying of cells with novel applications. In this Review, we present the physical principles of microscale EP devices and examine design trends in recent years. In addition, we discuss the use of reversible and irreversible EP in the development of therapeutics and analysis of intracellular contents, among other noteworthy applications. This Review aims to inform and encourage scientists and engineers to expand the use of efficient and versatile microscale EP technologies.
Collapse
Affiliation(s)
- Sung-Eun Choi
- Department of Mechanical Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, Maryland 21218, United States
| | - Harrison Khoo
- Department of Mechanical Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, Maryland 21218, United States
| | - Soojung Claire Hur
- Department of Mechanical Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, Maryland 21218, United States
- Institute for NanoBioTechnology, Johns Hopkins University, 3400 North Charles Street, Baltimore, Maryland 21218, United States
- Department of Oncology, Johns Hopkins University, 3400 North Charles Street, Baltimore, Maryland 21218, United States
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, 401 North Broadway, Baltimore, Maryland 21231, United States
| |
Collapse
|
14
|
Fang J, Huang S, Liu F, He G, Li X, Huang X, Chen HJ, Xie X. Semi-Implantable Bioelectronics. NANO-MICRO LETTERS 2022; 14:125. [PMID: 35633391 PMCID: PMC9148344 DOI: 10.1007/s40820-022-00818-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/09/2022] [Indexed: 06/15/2023]
Abstract
Developing techniques to effectively and real-time monitor and regulate the interior environment of biological objects is significantly important for many biomedical engineering and scientific applications, including drug delivery, electrophysiological recording and regulation of intracellular activities. Semi-implantable bioelectronics is currently a hot spot in biomedical engineering research area, because it not only meets the increasing technical demands for precise detection or regulation of biological activities, but also provides a desirable platform for externally incorporating complex functionalities and electronic integration. Although there is less definition and summary to distinguish it from the well-reviewed non-invasive bioelectronics and fully implantable bioelectronics, semi-implantable bioelectronics have emerged as highly unique technology to boost the development of biochips and smart wearable device. Here, we reviewed the recent progress in this field and raised the concept of "Semi-implantable bioelectronics", summarizing the principle and strategies of semi-implantable device for cell applications and in vivo applications, discussing the typical methodologies to access to intracellular environment or in vivo environment, biosafety aspects and typical applications. This review is meaningful for understanding in-depth the design principles, materials fabrication techniques, device integration processes, cell/tissue penetration methodologies, biosafety aspects, and applications strategies that are essential to the development of future minimally invasive bioelectronics.
Collapse
Affiliation(s)
- 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, People's Republic of 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, People's Republic of China
| | - Fanmao 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, People's Republic of China
| | - 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, People's Republic of China
| | - Xiangling Li
- 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, People's Republic of 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, People's Republic of 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, People's Republic of 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, People's Republic of China.
| |
Collapse
|
15
|
Zhang A, Fang J, Li X, Wang J, Chen M, Chen HJ, He G, Xie X. Cellular nanointerface of vertical nanostructure arrays and its applications. NANOSCALE ADVANCES 2022; 4:1844-1867. [PMID: 36133409 PMCID: PMC9419580 DOI: 10.1039/d1na00775k] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 11/28/2021] [Indexed: 06/16/2023]
Abstract
Vertically standing nanostructures with various morphologies have been developed with the emergence of the micro-/nanofabrication technology. When cells are cultured on them, various bio-nano interfaces between cells and vertical nanostructures would impact the cellular activities, depending on the shape, density, and height of nanostructures. Many cellular pathway activation processes involving a series of intracellular molecules (proteins, RNA, DNA, enzymes, etc.) would be triggered by the cell morphological changes induced by nanostructures, affecting the cell proliferation, apoptosis, differentiation, immune activation, cell adhesion, cell migration, and other behaviors. In addition, the highly localized cellular nanointerface enhances coupled stimulation on cells. Therefore, understanding the mechanism of the cellular nanointerface can not only provide innovative tools for regulating specific cell functions but also offers new aspects to understand the fundamental cellular activities that could facilitate the precise monitoring and treatment of diseases in the future. This review mainly describes the fabrication technology of vertical nanostructures, analyzing the formation of cellular nanointerfaces and the effects of cellular nanointerfaces on cells' fates and functions. At last, the applications of cellular nanointerfaces based on various nanostructures are summarized.
Collapse
Affiliation(s)
- Aihua Zhang
- State Key Laboratory of Optoelectronic Materials and Technologies, Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University Guangzhou 510006 Guangdong Province China
| | - Jiaru Fang
- State Key Laboratory of Optoelectronic Materials and Technologies, Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University Guangzhou 510006 Guangdong Province China
| | - Xiangling Li
- State Key Laboratory of Optoelectronic Materials and Technologies, Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University Guangzhou 510006 Guangdong Province China
- School of Biomedical Engineering, Sun Yat-Sen University Guangzhou 510006 China
| | - Ji Wang
- The First Affiliated Hospital of Sun Yat-Sen University Guangzhou 510080 China
| | - Meiwan Chen
- Institute of Chinese Medical Sciences, University of Macau Taipa Macau SAR China
| | - Hui-Jiuan Chen
- State Key Laboratory of Optoelectronic Materials and Technologies, Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University Guangzhou 510006 Guangdong Province China
| | - Gen He
- State Key Laboratory of Optoelectronic Materials and Technologies, Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University Guangzhou 510006 Guangdong Province China
- Key Laboratory of Molecular Target & Clinical Pharmacology, State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University Guangzhou 511436 P. R. China
| | - Xi Xie
- State Key Laboratory of Optoelectronic Materials and Technologies, Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University Guangzhou 510006 Guangdong Province China
- The First Affiliated Hospital of Sun Yat-Sen University Guangzhou 510080 China
| |
Collapse
|
16
|
Mariano A, Lubrano C, Bruno U, Ausilio C, Dinger NB, Santoro F. Advances in Cell-Conductive Polymer Biointerfaces and Role of the Plasma Membrane. Chem Rev 2022; 122:4552-4580. [PMID: 34582168 PMCID: PMC8874911 DOI: 10.1021/acs.chemrev.1c00363] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Indexed: 02/07/2023]
Abstract
The plasma membrane (PM) is often described as a wall, a physical barrier separating the cell cytoplasm from the extracellular matrix (ECM). Yet, this wall is a highly dynamic structure that can stretch, bend, and bud, allowing cells to respond and adapt to their surrounding environment. Inspired by shapes and geometries found in the biological world and exploiting the intrinsic properties of conductive polymers (CPs), several biomimetic strategies based on substrate dimensionality have been tailored in order to optimize the cell-chip coupling. Furthermore, device biofunctionalization through the use of ECM proteins or lipid bilayers have proven successful approaches to further maximize interfacial interactions. As the bio-electronic field aims at narrowing the gap between the electronic and the biological world, the possibility of effectively disguising conductive materials to "trick" cells to recognize artificial devices as part of their biological environment is a promising approach on the road to the seamless platform integration with cells.
Collapse
Affiliation(s)
- Anna Mariano
- Tissue
Electronics, Istituto Italiano di Tecnologia, 80125 Naples, Italy
| | - Claudia Lubrano
- Tissue
Electronics, Istituto Italiano di Tecnologia, 80125 Naples, Italy
- Dipartimento
di Chimica, Materiali e Produzione Industriale, Università di Napoli Federico II, 80125 Naples, Italy
| | - Ugo Bruno
- Tissue
Electronics, Istituto Italiano di Tecnologia, 80125 Naples, Italy
- Dipartimento
di Chimica, Materiali e Produzione Industriale, Università di Napoli Federico II, 80125 Naples, Italy
| | - Chiara Ausilio
- Tissue
Electronics, Istituto Italiano di Tecnologia, 80125 Naples, Italy
| | - Nikita Bhupesh Dinger
- Tissue
Electronics, Istituto Italiano di Tecnologia, 80125 Naples, Italy
- Dipartimento
di Chimica, Materiali e Produzione Industriale, Università di Napoli Federico II, 80125 Naples, Italy
| | - Francesca Santoro
- Tissue
Electronics, Istituto Italiano di Tecnologia, 80125 Naples, Italy
| |
Collapse
|
17
|
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: 7.0] [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
|
18
|
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: 38] [Impact Index Per Article: 12.7] [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
|
19
|
Maschietto M, Dal Maschio M, Girardi S, Vassanelli S. In situ electroporation of mammalian cells through SiO 2 thin film capacitive microelectrodes. Sci Rep 2021; 11:15126. [PMID: 34302040 PMCID: PMC8302607 DOI: 10.1038/s41598-021-94620-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 07/05/2021] [Indexed: 11/19/2022] Open
Abstract
Electroporation is a widely used non-viral technique for the delivery of molecules, including nucleic acids, into cells. Recently, electronic microsystems that miniaturize the electroporation machinery have been developed as a new tool for genetic manipulation of cells in vitro, by integrating metal microelectrodes in the culture substrate and enabling electroporation in-situ. We report that non-faradic SiO2 thin film-insulated microelectrodes can be used for reliable and spatially selective in-situ electroporation of mammalian cells. CHO-K1 and SH-SY5Y cell lines and primary neuronal cultures were electroporated by application of short and low amplitude voltage transients leading to cell electroporation by capacitive currents. We demonstrate reliable delivery of DNA plasmids and exogenous gene expression, accompanied by high spatial selectivity and cell viability, even with differentiated neurons. Finally, we show that SiO2 thin film-insulated microelectrodes support a double and serial transfection of the targeted cells.
Collapse
Affiliation(s)
- M Maschietto
- Department of Biomedical Sciences, Section of Physiology, University of Padua, via F. Marzolo 3, 35131, Padua, Italy
| | - M Dal Maschio
- Department of Biomedical Sciences, Section of Physiology, University of Padua, via F. Marzolo 3, 35131, Padua, Italy
| | - S Girardi
- Department of Biomedical Sciences, Section of Physiology, University of Padua, via F. Marzolo 3, 35131, Padua, Italy
| | - S Vassanelli
- Department of Biomedical Sciences, Section of Physiology, University of Padua, via F. Marzolo 3, 35131, Padua, Italy. .,Padua Neuroscience Center, University of Padua, via Orus 2/B, 35131, Padua, Italy. .,Institute of Condensed Matter Chemistry and Technologies for Energy, CNR, Corso Stati Uniti 4, 35127, Padua, Italy.
| |
Collapse
|
20
|
Spiteri C, Caprettini V, Chiappini C. Biomaterials-based approaches to model embryogenesis. Biomater Sci 2021; 8:6992-7013. [PMID: 33136109 DOI: 10.1039/d0bm01485k] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Understanding, reproducing, and regulating the cellular and molecular processes underlying human embryogenesis is critical to improve our ability to recapitulate tissues with proper architecture and function, and to address the dysregulation of embryonic programs that underlies birth defects and cancer. The rapid emergence of stem cell technologies is enabling enormous progress in understanding embryogenesis using simple, powerful, and accessible in vitro models. Biomaterials are playing a central role in providing the spatiotemporal organisation of biophysical and biochemical signalling necessary to mimic, regulate and dissect the evolving embryonic niche in vitro. This contribution is rapidly improving our understanding of the mechanisms underlying embryonic patterning, in turn enabling the development of more effective clinical interventions for regenerative medicine and oncology. Here we highlight how key biomaterial approaches contribute to organise signalling in human embryogenesis models, and we summarise the biological insights gained from these contributions. Importantly, we highlight how nanotechnology approaches have remained largely untapped in this space, and we identify their key potential contributions.
Collapse
Affiliation(s)
- Chantelle Spiteri
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK.
| | | | | |
Collapse
|
21
|
Sero JE, Stevens MM. Nanoneedle-Based Materials for Intracellular Studies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1295:191-219. [PMID: 33543461 DOI: 10.1007/978-3-030-58174-9_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Nanoneedles, defined as high aspect ratio structures with tip diameters of 5 to approximately 500 nm, are uniquely able to interface with the interior of living cells. Their nanoscale dimensions mean that they are able to penetrate the plasma membrane with minimal disruption of normal cellular functions, allowing researchers to probe the intracellular space and deliver or extract material from individual cells. In the last decade, a variety of strategies have been developed using nanoneedles, either singly or as arrays, to investigate the biology of cancer cells in vitro and in vivo. These include hollow nanoneedles for soluble probe delivery, nanocapillaries for single-cell biopsy, nano-AFM for direct physical measurements of cytosolic proteins, and a wide range of fluorescent and electrochemical nanosensors for analyte detection. Nanofabrication has improved to the point that nanobiosensors can detect individual vesicles inside the cytoplasm, delineate tumor margins based on intracellular enzyme activity, and measure changes in cell metabolism almost in real time. While most of these applications are currently in the proof-of-concept stage, nanoneedle technology is poised to offer cancer biologists a powerful new set of tools for probing cells with unprecedented spatial and temporal resolution.
Collapse
Affiliation(s)
- Julia E Sero
- Biology and Biochemistry Department, University of Bath, Claverton Down, Bath, UK
| | - Molly M Stevens
- Institute for Biomedical Engineering, Imperial College London, London, UK.
| |
Collapse
|
22
|
Brooks J, Minnick G, Mukherjee P, Jaberi A, Chang L, Espinosa HD, Yang R. High Throughput and Highly Controllable Methods for In Vitro Intracellular Delivery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2004917. [PMID: 33241661 PMCID: PMC8729875 DOI: 10.1002/smll.202004917] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/06/2020] [Indexed: 05/03/2023]
Abstract
In vitro and ex vivo intracellular delivery methods hold the key for releasing the full potential of tissue engineering, drug development, and many other applications. In recent years, there has been significant progress in the design and implementation of intracellular delivery systems capable of delivery at the same scale as viral transfection and bulk electroporation but offering fewer adverse outcomes. This review strives to examine a variety of methods for in vitro and ex vivo intracellular delivery such as flow-through microfluidics, engineered substrates, and automated probe-based systems from the perspective of throughput and control. Special attention is paid to a particularly promising method of electroporation using micro/nanochannel based porous substrates, which expose small patches of cell membrane to permeabilizing electric field. Porous substrate electroporation parameters discussed include system design, cells and cargos used, transfection efficiency and cell viability, and the electric field and its effects on molecular transport. The review concludes with discussion of potential new innovations which can arise from specific aspects of porous substrate-based electroporation platforms and high throughput, high control methods in general.
Collapse
Affiliation(s)
- Justin Brooks
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Grayson Minnick
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Prithvijit Mukherjee
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Arian Jaberi
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Lingqian Chang
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Horacio D. Espinosa
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Theoretical and Applied Mechanics Program, Northwestern University, Evanston, IL, 60208, USA
| | - Ruiguo Yang
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| |
Collapse
|
23
|
Duckert B, Vinkx S, Braeken D, Fauvart M. Single-cell transfection technologies for cell therapies and gene editing. J Control Release 2020; 330:963-975. [PMID: 33160005 DOI: 10.1016/j.jconrel.2020.10.068] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/30/2020] [Accepted: 10/31/2020] [Indexed: 12/29/2022]
Abstract
Advances in gene editing and cell therapies have recently led to outstanding clinical successes. However, the lack of a cost-effective manufacturing process prevents the democratization of these innovative medical tools. Due to the common use of viral vectors, the step of transfection in which cells are engineered to gain new functions, is a major bottleneck in making safe and affordable cell products. A promising opportunity lies in Single-Cell Transfection Technologies (SCTTs). SCTTs have demonstrated higher efficiency, safety and scalability than conventional transfection methods. They can also feature unique abilities such as substantial dosage control over the cargo delivery, single-cell addressability and integration in microdevices comprising multiple monitoring modalities. Unfortunately, the potential of SCTTs is not fully appreciated: they are most often restricted to research settings with little adoption in clinical settings. To encourage their adoption, we review and compare recent developments in SCTTs, and how they can enable selected clinical applications. To help bridge the gap between fundamental research and its translation to the clinic, we also describe how Good Manufacturing Practices (GMP) can be integrated in the design of SCTTs.
Collapse
Affiliation(s)
- Bastien Duckert
- Department of Physics and Astronomy, KU Leuven, Celestijnenlaan 200d, 3001 Leuven, Belgium; IMEC, Kapeldreef 75, 3001 Leuven, Belgium.
| | | | | | | |
Collapse
|
24
|
Bussooa A, Hoare D, Kirimi MT, Mitra S, Mirzai N, Neale SL, Mercer JR. Impedimetric Detection and Electromediated Apoptosis of Vascular Smooth Muscle Using Microfabricated Biosensors for Diagnosis and Therapeutic Intervention in Cardiovascular Diseases. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1902999. [PMID: 32999823 DOI: 10.1002/advs.2019029991902999] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/17/2020] [Indexed: 05/23/2023]
Abstract
Cardiovascular diseases remain a significant global burden with 1-in-3 of all deaths attributable to the consequences of the disease. The main cause is blocked arteries which often remain undetected. Implantable medical devices (IMDs) such as stents and grafts are often used to reopen vessels but over time these too will re-block. A vascular biosensor is developed that can report on cellularity and is amenable to being mounted on a stent or graft for remote reporting. Moreover, the device is designed to also receive currents that can induce a controlled form of cell death, apoptosis. A combined diagnostic and therapeutic biosensor would be transformational for the treatment of vascular diseases such as atherosclerosis and central line access. In this work, a cell sensing and cell apoptosing system based on the same interdigitated electrodes (IDEs) is developed. It is shown that the device is scalable and that by miniaturizing the IDEs, the detection sensitivity is increased. Apoptosis of vascular smooth muscle cells is monitored using continuous impedance measurements at a frequency of 10 kHz and rates of cell death are tracked using fluorescent dyes and live cell imaging.
Collapse
Affiliation(s)
- Anubhav Bussooa
- BHF Cardiovascular Research Centre University of Glasgow Glasgow G12 8TA UK
| | - Daniel Hoare
- BHF Cardiovascular Research Centre University of Glasgow Glasgow G12 8TA UK
| | - Mahmut T Kirimi
- BHF Cardiovascular Research Centre University of Glasgow Glasgow G12 8TA UK
| | - Srinjoy Mitra
- Scottish Microelectronics Centre Kings Buildings Campus University of Edinburgh Edinburgh EH9 3FF Scotland
| | - Nosrat Mirzai
- Bioelectronics Unit University of Glasgow Glasgow G12 8TA UK
| | - Steve L Neale
- James Watt School of Engineering University of Glasgow Glasgow G12 8QQ UK
| | - John R Mercer
- BHF Cardiovascular Research Centre University of Glasgow Glasgow G12 8TA UK
| |
Collapse
|
25
|
Bussooa A, Hoare D, Kirimi MT, Mitra S, Mirzai N, Neale SL, Mercer JR. Impedimetric Detection and Electromediated Apoptosis of Vascular Smooth Muscle Using Microfabricated Biosensors for Diagnosis and Therapeutic Intervention in Cardiovascular Diseases. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1902999. [PMID: 32999823 PMCID: PMC7509665 DOI: 10.1002/advs.201902999] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/17/2020] [Indexed: 05/23/2023]
Abstract
Cardiovascular diseases remain a significant global burden with 1-in-3 of all deaths attributable to the consequences of the disease. The main cause is blocked arteries which often remain undetected. Implantable medical devices (IMDs) such as stents and grafts are often used to reopen vessels but over time these too will re-block. A vascular biosensor is developed that can report on cellularity and is amenable to being mounted on a stent or graft for remote reporting. Moreover, the device is designed to also receive currents that can induce a controlled form of cell death, apoptosis. A combined diagnostic and therapeutic biosensor would be transformational for the treatment of vascular diseases such as atherosclerosis and central line access. In this work, a cell sensing and cell apoptosing system based on the same interdigitated electrodes (IDEs) is developed. It is shown that the device is scalable and that by miniaturizing the IDEs, the detection sensitivity is increased. Apoptosis of vascular smooth muscle cells is monitored using continuous impedance measurements at a frequency of 10 kHz and rates of cell death are tracked using fluorescent dyes and live cell imaging.
Collapse
Affiliation(s)
- Anubhav Bussooa
- BHF Cardiovascular Research CentreUniversity of GlasgowGlasgowG12 8TAUK
| | - Daniel Hoare
- BHF Cardiovascular Research CentreUniversity of GlasgowGlasgowG12 8TAUK
| | - Mahmut T. Kirimi
- BHF Cardiovascular Research CentreUniversity of GlasgowGlasgowG12 8TAUK
| | - Srinjoy Mitra
- Scottish Microelectronics CentreKings Buildings CampusUniversity of EdinburghEdinburgh EH9 3FFScotland
| | - Nosrat Mirzai
- Bioelectronics UnitUniversity of GlasgowGlasgowG12 8TAUK
| | - Steve L. Neale
- James Watt School of EngineeringUniversity of GlasgowGlasgowG12 8QQUK
| | - John R. Mercer
- BHF Cardiovascular Research CentreUniversity of GlasgowGlasgowG12 8TAUK
| |
Collapse
|
26
|
Shinde P, Kar S, Loganathan M, Chang HY, Tseng FG, Nagai M, Santra TS. Infrared Pulse Laser-Activated Highly Efficient Intracellular Delivery Using Titanium Microdish Device. ACS Biomater Sci Eng 2020; 6:5645-5652. [PMID: 33320577 DOI: 10.1021/acsbiomaterials.0c00785] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We report infrared (IR) pulse laser-activated highly efficient parallel intracellular delivery by using an array of titanium microdish (TMD) device. Upon IR laser pulse irradiation, a two-dimensional array of TMD device generated photothermal cavitation bubbles to disrupt the cell membrane surface and create transient membrane pores to deliver biomolecules into cells by a simple diffusion process. We successfully delivered the dyes and different sizes of dextran in different cell types with variations of laser pulses. Our platform has the ability to transfect more than a million cells in a parallel fashion within a minute. The best results were achieved for SiHa cells with a delivery efficiency of 96% and a cell viability of around 98% for propidium iodide dye using 600 pulses, whereas a delivery efficiency of 98% and a cell viability of 100% were obtained for dextran 3000 MW delivery using 700 pulses. For dextran 10,000 MW, the delivery efficiency was 92% and the cell viability was 98%, respectively. The device is compact, easy-to-use, and potentially applicable for cellular therapy and diagnostic purposes.
Collapse
Affiliation(s)
- Pallavi Shinde
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai 600036, India
| | - Srabani Kar
- Department of Electrical Engineering, University of Cambridge, Cambridge CB3 0FA U.K
| | - Mohan Loganathan
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai 600036, India.,Department of Mechanical Engineering, Toyohashi University of Technology, Toyohashi 441-8580, Japan
| | - Hwan-You Chang
- Department of Life Science, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Fan-Gang Tseng
- Department of Engineering and System Science, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Moeto Nagai
- Department of Mechanical Engineering, Toyohashi University of Technology, Toyohashi 441-8580, Japan
| | - Tuhin Subhra Santra
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai 600036, India
| |
Collapse
|
27
|
Tay A. The Benefits of Going Small: Nanostructures for Mammalian Cell Transfection. ACS NANO 2020; 14:7714-7721. [PMID: 32631053 DOI: 10.1021/acsnano.0c04624] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Nanostructures, with their localized interactions with mammalian cells, can offer better efficiency and lower cell perturbation than conventional viral, biochemical, and electroporation transfection techniques. In this Perspective, I describe the different stages of transfection and provide a comparison of transfection techniques based on their mechanisms. Focusing on specific aims of transfection, I also illustrate how recent developments in high-aspect-ratio nanostructures have endowed them with properties that are superior to existing viral, biochemical, and electroporation methods as a versatile technique to deliver a variety of cargoes and to interface with different mammalian cell types for biomedical applications. Finally, I describe the challenges associated with transfection that need to be overcome to enhance cargo delivery efficiency and clinical translation.
Collapse
Affiliation(s)
- Andy Tay
- Department of Biomedical Engineering, National University of Singapore, Singapore 117583
| |
Collapse
|
28
|
Bruno G, Colistra N, Melle G, Cerea A, Hubarevich A, Deleye L, De Angelis F, Dipalo M. Microfluidic Multielectrode Arrays for Spatially Localized Drug Delivery and Electrical Recordings of Primary Neuronal Cultures. Front Bioeng Biotechnol 2020; 8:626. [PMID: 32656200 PMCID: PMC7325920 DOI: 10.3389/fbioe.2020.00626] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 05/21/2020] [Indexed: 12/15/2022] Open
Abstract
Neuropathological models and neurological disease progression and treatments have always been of great interest in biomedical research because of their impact on society. The application of in vitro microfluidic devices to neuroscience-related disciplines provided several advancements in therapeutics or neuronal modeling thanks to the ability to control the cellular microenvironment at spatiotemporal level. Recently, the introduction of three-dimensional nanostructures has allowed high performance in both in vitro recording of electrogenic cells and drug delivery using minimally invasive devices. Independently, both delivery and recording have let to pioneering solutions in neurobiology. However, their combination on a single chip would provide further fundamental improvements in drug screening systems and would offer comprehensive insights into pathologies and diseases progression. Therefore, it is crucial to develop platforms able to monitor progressive changes in electrophysiological behavior in the electrogenic cellular network, induced by spatially localized injection of biochemical agents. In this work, we show the application of a microfluidic multielectrode array (MEA) platform to record spontaneous and chemically stimulated activity in primary neuronal networks. By means of spatially localized caffeine injection via microfluidic nanochannels, the device demonstrated its capability of combined localized drug delivery and cell signaling recording. The platform could detect activity of the neural network at multiple sites while delivering molecules into just a few selected cells, thereby examining the effect of biochemical agents on the desired portion of cell culture.
Collapse
Affiliation(s)
- Giulia Bruno
- DIBRIS, Università degli Studi di Genova, Genoa, Italy.,Istituto Italiano di Tecnologia, Genoa, Italy
| | | | - Giovanni Melle
- DIBRIS, Università degli Studi di Genova, Genoa, Italy.,Istituto Italiano di Tecnologia, Genoa, Italy
| | | | | | | | | | | |
Collapse
|
29
|
Recent advances in micro/nanoscale intracellular delivery. NANOTECHNOLOGY AND PRECISION ENGINEERING 2020. [DOI: 10.1016/j.npe.2019.12.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
30
|
Wu Y, Chen H, Guo L. Opportunities and dilemmas of in vitro nano neural electrodes. RSC Adv 2020; 10:187-200. [PMID: 35492533 PMCID: PMC9047985 DOI: 10.1039/c9ra08917a] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 12/04/2019] [Indexed: 01/05/2023] Open
Abstract
Developing electrophysiological platforms to capture electrical activities of neurons and exert modulatory stimuli lays the foundation for many neuroscience-related disciplines, including the neuron–machine interface, neuroprosthesis, and mapping of brain circuitry.
Collapse
Affiliation(s)
- Yu Wu
- Department of Electrical and Computer Engineering
- The Ohio State University
- Columbus
- USA
| | - Haowen Chen
- Department of Electrical and Computer Engineering
- The Ohio State University
- Columbus
- USA
| | - Liang Guo
- Department of Electrical and Computer Engineering
- The Ohio State University
- Columbus
- USA
| |
Collapse
|
31
|
Wen R, Zhang AH, Liu D, Feng J, Yang J, Xia D, Wang J, Li C, Zhang T, Hu N, Hang T, He G, Xie X. Intracellular Delivery and Sensing System Based on Electroplated Conductive Nanostraw Arrays. ACS APPLIED MATERIALS & INTERFACES 2019; 11:43936-43948. [PMID: 31696695 DOI: 10.1021/acsami.9b15619] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
One-dimensional nanoneedle-like arrays have emerged as an attractive tool for penetrating the cell membrane to achieve intracellular applications including drug delivery, electrical recording, and biochemical detection. Hollow nanoneedles, also called nanostraws (NSs), combined with nanoelectroporation have been demonstrated as a powerful platform for intracellular drug delivery and extraction of intracellular contents. However, the fabrication technique of nanostraws still requires complicated and expensive atomic layer deposition and etching processes and fails to produce conductive nanostraws. Herein, we developed a commonly accessible and versatile electrodeposition approach to controllably fabricate conductive nanostraw arrays based on various types of metal or conductive polymer materials. Representatively, Pt nanostraws (Pt NSs) with 400 nm diameter were further integrated with a low-voltage nanoelectroporation system to achieve cell detection, intracellular drug delivery, and sensing of intracellular enzymes. Both theoretical simulations and experimental results revealed that the conductive nanostraws in direct contact with cells could induce high-efficiency cell electroporation at relatively low voltage (∼5 V). Efficient delivery of reagents into live cells with spatial control and repeated extraction of intracellular enzymes (e.g., caspase-3) for temporal monitoring from the same set of cells were demonstrated. This work not only pioneers a new avenue for universal production of conductive nanostraws on a large scale but also presents great potential for developing nanodevices to achieve a variety of biomedical applications including cell re-engineering, cell-based therapy, and signaling pathway monitoring.
Collapse
Affiliation(s)
- Rui Wen
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, The First Affiliated Hospital of Sun Yat-Sen University , Sun Yat-Sen University , Guangzhou 510006 , China
| | - Ai-Hua Zhang
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, The First Affiliated Hospital of Sun Yat-Sen University , Sun Yat-Sen University , Guangzhou 510006 , China
| | - Di Liu
- Pritzker School of Medicine , University of Chicago , Chicago , Illinois 60637 , United States
| | - Jianming Feng
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, The First Affiliated Hospital of Sun Yat-Sen University , Sun Yat-Sen University , Guangzhou 510006 , 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 , China
| | - Dehua Xia
- School of Environmental Science and Engineering , Sun Yat-sen University , Guangzhou 510275 , China
| | - Ji Wang
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, The First Affiliated Hospital of Sun Yat-Sen University , Sun Yat-Sen University , Guangzhou 510006 , China
| | - Chunwei Li
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, The First Affiliated Hospital of Sun Yat-Sen University , Sun Yat-Sen University , Guangzhou 510006 , China
| | - Tao Zhang
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, The First Affiliated Hospital of Sun Yat-Sen University , Sun Yat-Sen University , Guangzhou 510006 , China
| | - Ning Hu
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, The First Affiliated Hospital of Sun Yat-Sen University , Sun Yat-Sen University , Guangzhou 510006 , China
| | - Tian Hang
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, The First Affiliated Hospital of Sun Yat-Sen University , Sun Yat-Sen University , Guangzhou 510006 , China
| | - 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, The First Affiliated Hospital of Sun Yat-Sen University , Sun Yat-Sen University , Guangzhou 510006 , 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, The First Affiliated Hospital of Sun Yat-Sen University , Sun Yat-Sen University , Guangzhou 510006 , China
| |
Collapse
|
32
|
Dipalo M, Caprettini V, Bruno G, Caliendo F, Garma LD, Melle G, Dukhinova M, Siciliano V, Santoro F, De Angelis F. Membrane Poration Mechanisms at the Cell-Nanostructure Interface. ACTA ACUST UNITED AC 2019; 3:e1900148. [PMID: 32648684 DOI: 10.1002/adbi.201900148] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/21/2019] [Indexed: 01/27/2023]
Abstract
3D vertical nanostructures have become one of the most significant methods for interfacing cells and the nanoscale and for accessing significant intracellular functionalities such as membrane potential. As this intracellular access can be induced by means of diverse cellular membrane poration mechanisms, it is important to investigate in detail the cell condition after membrane rupture for assessing the real effects of the poration techniques on the biological environment. Indeed, differences of the membrane dynamics and reshaping have not been observed yet when the membrane-nanostructure system is locally perturbed by, for instance, diverse membrane breakage events. In this work, new insights are provided into the membrane dynamics in case of two different poration approaches, optoacoustic- and electro-poration, both mediated by the same 3D nanostructures. The experimental results offer a detailed overview on the different poration processes in terms of electrical recordings and membrane conformation.
Collapse
Affiliation(s)
| | | | - Giulia Bruno
- Istituto Italiano di Tecnologia, Genoa, 16163, Italy
- Dipartimento di Informatica, Bioingegneria, Robotica e Ingegneria dei Sistemi. DIBRIS, Università degli Studi di Genova, Genova, 16126, Italy
| | - Fabio Caliendo
- Center for Advacend Biomaterials for Healthcare, Istituto Italiano di Tecnologia, Naples, 80125, Italy
| | - Leonardo D Garma
- Center for Advacend Biomaterials for Healthcare, Istituto Italiano di Tecnologia, Naples, 80125, Italy
| | - Giovanni Melle
- Istituto Italiano di Tecnologia, Genoa, 16163, Italy
- Dipartimento di Informatica, Bioingegneria, Robotica e Ingegneria dei Sistemi. DIBRIS, Università degli Studi di Genova, Genova, 16126, Italy
| | - Marina Dukhinova
- Center for Advacend Biomaterials for Healthcare, Istituto Italiano di Tecnologia, Naples, 80125, Italy
| | - Velia Siciliano
- Center for Advacend Biomaterials for Healthcare, Istituto Italiano di Tecnologia, Naples, 80125, Italy
| | - Francesca Santoro
- Center for Advacend Biomaterials for Healthcare, Istituto Italiano di Tecnologia, Naples, 80125, Italy
| | | |
Collapse
|
33
|
Effect of oblique polymer pillars on spreading and elongation of rat mesenchymal stem cells. Colloids Surf B Biointerfaces 2019; 183:110485. [PMID: 31499453 DOI: 10.1016/j.colsurfb.2019.110485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 08/28/2019] [Accepted: 09/01/2019] [Indexed: 11/23/2022]
Abstract
Stiffness and anisotropy of culture substrates are important factors influencing the cell behavior and their responses to external stimuli. Herein, we report a fabrication method of oblique polymer pillars which allow modulating both stiffness and anisotropy of the substrate for spreading and elongation studies of Rat Mesenchymal Stem Cells (RMSCs). Poly (Lactic-co-Glycolic Acid) (PLGA) has been chosen to produce micro-pillars of different heights and different pitches using a combined method of soft-lithography and hot embossing. The stiffness of such pillar substrates varies over a large range so that RMSCs show effectively different spreading behaviors which are also sensitive to the inclining angle of the pillars. Our results showed that with the increase of the pillar height the area of cell spreading decreases but the cell elongation aspect ratio increases. Moreover, cells preferentially elongate along the direction perpendicular to that of the pillars' inclining, which is in agreement with the calculated anisotropy of the pillar substrate stiffness.
Collapse
|
34
|
Abstract
Intracellular cargo delivery is an essential step in many biomedical applications including gene editing and biologics therapy. Examples of cargo include nucleic acids (RNA and DNA), proteins, small biomolecules, and drugs, which can vary substantially in terms of their sizes, charges, solubility, and stability. Viruses have been used traditionally to deliver nucleic acids into cells, but the method suffers from limitations such as small cargo size, safety concerns, and viral genome integration into host cells, all of which complicate therapeutic applications. Commercially available techniques using biochemicals and bulk electroporation are, in general, poorly compatible with primary cells such as human induced pluripotent stem cells and immune cells, which are increasingly important candidates for adoptive cell therapy. Nanostructures, with dimensions ranging from tens of nanometers to a few micrometers, may play a critical role in overcoming cellular manipulation and delivery challenges and provide a powerful alternative to conventional techniques. A critical feature that differentiates nanostructures from viral, biochemical, and bulk electroporation techniques is that they interface with cells at a scale measuring ten to hundreds of nanometers in size. This highly local interaction enables application of stronger and more direct stimuli such as mechanical force, heat, or electric fields than would be possible in a bulk treatment. Compared to popular viral, biochemical, and bulk electroporation methods, nanostructures were found to minimally perturb cells with cells remaining in good health during postdelivery culture. These advantages have enabled nanostructures such as nanowires and nanotubes to successfully interface with a wide variety of cells, including primary immune cells and cardiomyocytes, for in vitro and in vivo applications. This Account is focused on using nanostructures for cargo delivery into biological cells. In this Account, we will first outline the historical developments using nanostructures for interfacing with cells. We will highlight how mechanistic understanding of nano-bio interactions has evolved over the last decade and how this improved knowledge has motivated coupling of electric and magnetic fields to nanostructures to improve delivery outcomes. There will also be an in-depth discussion on the merits of nanostructures in comparison to conventional methods using viruses, biochemicals, and bulk electroporation. Finally, motivated by our observations on the lack of consistency in reporting key metrics such as efficiency in literature, we suggest a set of metrics for documenting experimental results with the aim to promote standardization in reporting and ease in comparing. We suggest the use of more sophisticated tools such as RNA transcriptomics for thorough assessment of cell perturbation attributed to intracellular delivery. We hope that this Account can effectively capture the progress of nanostructure-mediated cargo delivery and encourage new innovations.
Collapse
Affiliation(s)
- Andy Tay
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States,Department of Biomedical Engineering, National University of Singapore, 117583 Singapore
| | - Nicholas Melosh
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States,Corresponding Author:
| |
Collapse
|
35
|
Soft Electroporation Through 3D Hollow Nanoelectrodes. Methods Mol Biol 2019. [PMID: 31468475 DOI: 10.1007/978-1-4939-9740-4_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Generally, electroporation of in vitro cells is performed under very high electric fields to overcome the physical barrier of plasma membrane. Since traditional electroporation techniques make use of very high voltages, which is critical to cell viability, this study presents a microfluidic platform able to perform cell membrane electroporation with the application of low voltages (1.5-2 V). The platform is manufactured based on the milling by mean of focused ionic beam, which offers an established approach to fabricate ordered arrays of 3D gold hollow nanoelectrodes protruding from an insulating substrate. The novelty of this fabrication relies on the fact that the nanoelectrodes used for electroporation are simultaneously metallic, hollow and communicate through its nanochannels with an isolated microfluidic chamber beneath the device. Adherent cultured cells on the nanoelectrodes can be electroporated in this platform, and molecules can be selectively delivered only inside the porated cells.
Collapse
|
36
|
Abstract
Nanostructured devices are able to foster the technology for cell membrane poration. With the size smaller than a cell, nanostructures allow efficient poration on the cell membrane. Emerging nanostructures with various physical transduction have been demonstrated to accommodate effective intracellular delivery. Aside from improving poration and intracellular delivery performance, nanostructured devices also allow for the discovery of novel physiochemical phenomena and the biological response of the cell. This article provides a brief introduction to the principles of nanostructured devices for cell poration and outlines the intracellular delivery capability of the technology. In the future, we envision more exploration on new nanostructure designs and creative applications in biomedical fields.
Collapse
Affiliation(s)
- Apresio K Fajrial
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, 80309 United States of America
| | | |
Collapse
|
37
|
Nanostraw membrane stamping for direct delivery of molecules into adhesive cells. Sci Rep 2019; 9:6806. [PMID: 31048793 PMCID: PMC6497648 DOI: 10.1038/s41598-019-43340-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 04/18/2019] [Indexed: 01/23/2023] Open
Abstract
Delivering ions and molecules into living cells has become an important challenge in medical and biological fields. Conventional molecular delivery, however, has several issues such as physical and chemical damage to biological cells. Here, we present a method of directly delivering molecules into adhesive cells with an Au-based nanostraw membrane stamp that can physically inject a target molecule into the cytoplasm through a nanostraw duct. We successfully delivered calcein target molecules into adhesive cells with high efficiency (85%) and viability (90%). Furthermore, we modeled the molecular flow through Au nanostraws and then demonstrated the control of calcein flow by changing the concentration and geometry of Au nanostraws. Our Au membrane stamping provides a new way of accessing the cytoplasm to modulate cellular functions via injected molecules.
Collapse
|
38
|
Liu Z, Nie J, Miao B, Li J, Cui Y, Wang S, Zhang X, Zhao G, Deng Y, Wu Y, Li Z, Li L, Wang ZL. Self-Powered Intracellular Drug Delivery by a Biomechanical Energy-Driven Triboelectric Nanogenerator. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1807795. [PMID: 30721538 DOI: 10.1002/adma.201807795] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 01/13/2019] [Indexed: 06/09/2023]
Abstract
Nondestructive, high-efficiency, and on-demand intracellular drug/biomacromolecule delivery for therapeutic purposes remains a great challenge. Herein, a biomechanical-energy-powered triboelectric nanogenerator (TENG)-driven electroporation system is developed for intracellular drug delivery with high efficiency and minimal cell damage in vitro and in vivo. In the integrated system, a self-powered TENG as a stable voltage pulse source triggers the increase of plasma membrane potential and membrane permeability. Cooperatively, the silicon nanoneedle-array electrode minimizes cellular damage during electroporation via enhancing the localized electrical field at the nanoneedle-cell interface and also decreases plasma membrane fluidity for the enhancement of molecular influx. The integrated system achieves efficient delivery of exogenous materials (small molecules, macromolecules, and siRNA) into different types of cells, including hard-to-transfect primary cells, with delivery efficiency up to 90% and cell viability over 94%. Through simple finger friction or hand slapping of the wearable TENGs, it successfully realizes a transdermal biomolecule delivery with an over threefold depth enhancement in mice. This integrated and self-powered system for active electroporation drug delivery shows great prospect for self-tuning drug delivery and wearable medicine.
Collapse
Affiliation(s)
- Zhirong Liu
- Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience, Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 100083, P. R. China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jinhui Nie
- Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience, Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 100083, P. R. China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Bin Miao
- i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215125, P. R. China
| | - Jiadong Li
- i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215125, P. R. China
- State Key Laboratory of Applied Optics, Changchun Institute of Optics Fine Mechanics and Physics, Chinese Academy of Sciences, Changchun, 130033, P. R. China
| | - Yuanbo Cui
- Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Shu Wang
- Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience, Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 100083, P. R. China
| | - Xiaodi Zhang
- Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience, Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 100083, P. R. China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Gengrui Zhao
- Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience, Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 100083, P. R. China
| | - Yongbo Deng
- State Key Laboratory of Applied Optics, Changchun Institute of Optics Fine Mechanics and Physics, Chinese Academy of Sciences, Changchun, 130033, P. R. China
| | - Yihui Wu
- State Key Laboratory of Applied Optics, Changchun Institute of Optics Fine Mechanics and Physics, Chinese Academy of Sciences, Changchun, 130033, P. R. China
| | - Zhou Li
- Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience, Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 100083, P. R. China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- Center of Nanoenergy Research, School of Physical Science and Technology, Guangxi University, Nanning, 530004, P. R. China
| | - Linlin Li
- Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience, Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 100083, P. R. China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- Center of Nanoenergy Research, School of Physical Science and Technology, Guangxi University, Nanning, 530004, P. R. China
| | - Zhong Lin Wang
- Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience, Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 100083, P. R. China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- Center of Nanoenergy Research, School of Physical Science and Technology, Guangxi University, Nanning, 530004, P. R. China
- School of Materials Science and Engineering, Georgia Institute of Technology, Atlanta, GA, 30332-0245, USA
| |
Collapse
|
39
|
Huang JA, Caprettini V, Zhao Y, Melle G, Maccaferri N, Deleye L, Zambrana-Puyalto X, Ardini M, Tantussi F, Dipalo M, De Angelis F. On-Demand Intracellular Delivery of Single Particles in Single Cells by 3D Hollow Nanoelectrodes. NANO LETTERS 2019; 19:722-731. [PMID: 30673248 PMCID: PMC6378653 DOI: 10.1021/acs.nanolett.8b03764] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Delivery of molecules into intracellular compartments is one of the fundamental requirements in molecular biology. However, the possibility of delivering a precise number of nano-objects with single-particle resolution is still an open challenge. Here we present an electrophoretic platform based on 3D hollow nanoelectrodes to enable delivery of single nanoparticles into single selected cells and monitoring of the single-particle delivery by surface-enhanced Raman scattering (SERS). The gold-coated hollow nanoelectrode capable of confinement and enhancement of electromagnetic fields upon laser illumination can distinguish the SERS signals of a single nanoparticle flowing through the nanoelectrode. Tight wrapping of cell membranes around the nanoelectrodes allows effective membrane electroporation such that single gold nanorods are delivered on demand into a living cell by electrophoresis. The capability of the 3D hollow nanoelectrodes to porate cells and reveal single emitters from the background in continuous flow is promising for the analysis of both intracellular delivery and sampling.
Collapse
Affiliation(s)
- Jian-An Huang
- Istituto
Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Valeria Caprettini
- Istituto
Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
- DIBRIS, University of Genoa, Via all’Opera Pia 13, 16145 Genova, Italy
| | - Yingqi Zhao
- Istituto
Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Giovanni Melle
- Istituto
Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
- DIBRIS, University of Genoa, Via all’Opera Pia 13, 16145 Genova, Italy
| | | | - Lieselot Deleye
- Istituto
Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | | | - Matteo Ardini
- Istituto
Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | | | - Michele Dipalo
- Istituto
Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | | |
Collapse
|
40
|
Patchable micro/nanodevices interacting with skin. Biosens Bioelectron 2018; 122:189-204. [DOI: 10.1016/j.bios.2018.09.035] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 09/09/2018] [Accepted: 09/10/2018] [Indexed: 12/20/2022]
|
41
|
Caprettini V, Huang J, Moia F, Jacassi A, Gonano CA, Maccaferri N, Capozza R, Dipalo M, De Angelis F. Enhanced Raman Investigation of Cell Membrane and Intracellular Compounds by 3D Plasmonic Nanoelectrode Arrays. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2018; 5:1800560. [PMID: 30581692 PMCID: PMC6299714 DOI: 10.1002/advs.201800560] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 07/31/2018] [Indexed: 05/14/2023]
Abstract
3D nanostructures are widely exploited in cell cultures for many purposes such as controlled drug delivery, transfection, intracellular sampling, and electrical recording. However, little is known about the interaction of the cells with these substrates, and even less about the effects of electroporation on the cellular membrane and the nuclear envelope. This work exploits 3D plasmonic nanoelectrodes to study, by surface-enhanced Raman scattering (SERS), the cell membrane dynamics on the nanostructured substrate before, during, and after electroporation. In vitro cultured cells tightly adhere on 3D plasmonic nanoelectrodes precisely in the plasmonic hot spots, making this kind of investigation possible. After electroporation, the cell membrane dynamics are studied by recording the Raman time traces of biomolecules in contact or next to the 3D plasmonic nanoelectrode. During this process, the 3D plasmonic nanoelectrodes are intracellularly coupled, thus enabling the monitoring of different molecular species, including lipids, proteins, and nucleic acids. Scanning electron microscopy cross-section analysis evidences the possibility of nuclear membrane poration compatible with the reported Raman spectra. These findings may open a new route toward controlled intracellular sampling and intranuclear delivery of genic materials. They also show the possibility of nuclear envelope disruption which may lead to negative side effects.
Collapse
Affiliation(s)
| | - Jian‐An Huang
- Istituto Italiano di TecnologiaVia Morego 3016163GenoaItaly
| | - Fabio Moia
- Istituto Italiano di TecnologiaVia Morego 3016163GenoaItaly
| | - Andrea Jacassi
- Istituto Italiano di TecnologiaVia Morego 3016163GenoaItaly
| | | | | | | | - Michele Dipalo
- Istituto Italiano di TecnologiaVia Morego 3016163GenoaItaly
| | | |
Collapse
|
42
|
Cerea A, Caprettini V, Bruno G, Lovato L, Melle G, Tantussi F, Capozza R, Moia F, Dipalo M, De Angelis F. Selective intracellular delivery and intracellular recordings combined in MEA biosensors. LAB ON A CHIP 2018; 18:3492-3500. [PMID: 30306172 DOI: 10.1039/c8lc00435h] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Biological studies on in vitro cell cultures are of fundamental importance to investigate cell response to external stimuli, such as new drugs for the treatment of specific pathologies, or to study communication between electrogenic cells. Although three-dimensional (3D) nanostructures brought tremendous improvements on biosensors used for various biological in vitro studies, including drug delivery and electrical recording, there is still a lack of multifunctional capabilities that could help gain deeper insights in several bio-related research fields. In this work, the electrical recording of large cell ensembles and the intracellular delivery of few selected cells are combined on the same device by integrating microfluidic channels on the bottom of a multi-electrode array decorated with 3D hollow nanostructures. The novel platform allows the recording of intracellular-like action potentials from large ensembles of cardiomyocytes derived from human induced pluripotent stem cells (hiPSC) and from the HL-1 line, while different molecules are selectively delivered into single/few targeted cells. The proposed approach shows high potential for enabling new comprehensive studies that can relate drug effects to network level cell communication processes.
Collapse
Affiliation(s)
- Andrea Cerea
- Istituto Italiano di Tecnologia, 16163 Genova, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Mukaibo H. Template‐Synthesized Vertical Needle Array as Injection Platform for Microalgae. CHEM REC 2018; 19:859-872. [DOI: 10.1002/tcr.201800099] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 10/12/2018] [Indexed: 12/25/2022]
Affiliation(s)
- Hitomi Mukaibo
- Department of Chemical EngineeringUniversity of Rochester 4510 Wegmans Hall, Rochester, NY 14627 USA
| |
Collapse
|
44
|
Novickij V, Zinkevičienė A, Perminaitė E, Čėsna R, Lastauskienė E, Paškevičius A, Švedienė J, Markovskaja S, Novickij J, Girkontaitė I. Non-invasive nanosecond electroporation for biocontrol of surface infections: an in vivo study. Sci Rep 2018; 8:14516. [PMID: 30266920 PMCID: PMC6162327 DOI: 10.1038/s41598-018-32783-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 09/14/2018] [Indexed: 12/20/2022] Open
Abstract
Invasive infections caused by drug-resistant bacteria are frequently responsible for fatal sepsis, morbidity and mortality rates. In this work, we propose a new methodology based on nanosecond high frequency electric field bursts, which enables successful eradication of bacteria in vivo. High frequency (15 kHz) 15–25 kV/cm 300–900 ns pulsing bursts were used separately and in combination with acetic acid (0.1–1%) to treat Pseudomonas aeruginosa in a murine model. Acetic acid 1% alone was effective resulting in almost 10-fold reduction of bacteria viability, however combination of nanosecond electric field and acetic acid 1% treatment was the most successful showing almost full eradication (0.01% survival compared to control) of the bacteria in the contaminated area. The short duration of the pulses (sub-microsecond) and high frequency (kHz range) of the burst enabled reduction of the muscle contractions to barely detectable level while the proposed applicators ensured predominantly topical treatment, without electroporation of deeper tissues. The results of our study have direct application for treatment of wounds and ulcers when chemical treatment is no longer effective.
Collapse
Affiliation(s)
- Vitalij Novickij
- Institute of High Magnetic Fields, Vilnius Gediminas Technical University, Vilnius, Lithuania.
| | - Auksė Zinkevičienė
- State Research Institute Centre for Innovative Medicine, Department of Immunology, Vilnius, Lithuania
| | - Emilija Perminaitė
- State Research Institute Centre for Innovative Medicine, Department of Immunology, Vilnius, Lithuania
| | - Robertas Čėsna
- State Research Institute Centre for Innovative Medicine, Department of Immunology, Vilnius, Lithuania
| | - Eglė Lastauskienė
- Institute of Biosciences, Life Sciences Centre, Vilnius University, Vilnius, Lithuania
| | | | - Jurgita Švedienė
- Laboratory of Biodeterioration Research, Nature Research Centre, Vilnius, Lithuania
| | | | - Jurij Novickij
- Institute of High Magnetic Fields, Vilnius Gediminas Technical University, Vilnius, Lithuania
| | - Irutė Girkontaitė
- State Research Institute Centre for Innovative Medicine, Department of Immunology, Vilnius, Lithuania
| |
Collapse
|
45
|
Dipalot M, McGuiret AF, Lou HY, Caprettini V, Melle G, Bruno G, Lubrano C, Matino L, De Angelis F, Cui B, Santoro F. Cells Adhering to 3D Vertical Nanostructures: Cell Membrane Reshaping without Stable Internalization. NANO LETTERS 2018; 18:6100-6105. [PMID: 30091365 PMCID: PMC6485928 DOI: 10.1021/acs.nanolett.8b03163] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
The dynamic interface between the cellular membrane and 3D nanostructures determines biological processes and guides the design of novel biomedical devices. Despite the fact that recent advancements in the fabrication of artificial biointerfaces have yielded an enhanced understanding of this interface, there remain open questions on how the cellular membrane reacts and behaves in the presence of sharp objects on the nanoscale. Here we provide a multifaceted characterization of the cellular membrane's mechanical stability when closely interacting with high-aspect-ratio 3D vertical nanostructures, providing strong evidence that vertical nanostructures spontaneously penetrate the cellular membrane to form a steady intracellular coupling only in rare cases and under specific conditions. The cell membrane is able to conform tightly over the majority of structures with various shapes while maintaining its integrity.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Claudia Lubrano
- Center for Advanced Biomaterials for Healthcare, Istituto Italiano di Tecnologia, Napoli, Italy
| | - Laura Matino
- Center for Advanced Biomaterials for Healthcare, Istituto Italiano di Tecnologia, Napoli, Italy
| | | | | | - Francesca Santoro
- Center for Advanced Biomaterials for Healthcare, Istituto Italiano di Tecnologia, Napoli, Italy
| |
Collapse
|
46
|
Capozza R, Caprettini V, Gonano CA, Bosca A, Moia F, Santoro F, De Angelis F. Cell Membrane Disruption by Vertical Micro-/Nanopillars: Role of Membrane Bending and Traction Forces. ACS APPLIED MATERIALS & INTERFACES 2018; 10:29107-29114. [PMID: 30081625 PMCID: PMC6117743 DOI: 10.1021/acsami.8b08218] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Gaining access to the cell interior is fundamental for many applications, such as electrical recording and drug and biomolecular delivery. A very promising technique consists of culturing cells on micro-/nanopillars. The tight adhesion and high local deformation of cells in contact with nanostructures can promote the permeabilization of lipids at the plasma membrane, providing access to the internal compartment. However, there is still much experimental controversy regarding when and how the intracellular environment is targeted and the role of the geometry and interactions with surfaces. Consequently, we investigated, by coarse-grained molecular dynamics simulations of the cell membrane, the mechanical properties of the lipid bilayer under high strain and bending conditions. We found out that a high curvature of the lipid bilayer dramatically lowers the traction force necessary to achieve membrane rupture. Afterward, we experimentally studied the permeabilization rate of the cell membrane by pillars with comparable aspect ratios but different sharpness values at the edges. The experimental data support the simulation results: even pillars with diameters in the micron range may cause local membrane disruption when their edges are sufficiently sharp. Therefore, the permeabilization likelihood is connected to the local geometric features of the pillars rather than diameter or aspect ratio. The present study can also provide significant contributions to the design of three-dimensional biointerfaces for tissue engineering and cellular growth.
Collapse
Affiliation(s)
- Rosario Capozza
- Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Valeria Caprettini
- Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
- Università
degli studi di Genova, Genova 16126, Italy
| | - Carlo A. Gonano
- Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Alessandro Bosca
- Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Fabio Moia
- Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Francesca Santoro
- Center
for Advanced Biomaterials for Healthcare, Istituto Italiano di Tecnologia, 80125 Napoli, Italy
| | | |
Collapse
|
47
|
Kavaldzhiev MN, Perez JE, Sougrat R, Bergam P, Ravasi T, Kosel J. Inductively actuated micro needles for on-demand intracellular delivery. Sci Rep 2018; 8:9918. [PMID: 29967360 PMCID: PMC6028653 DOI: 10.1038/s41598-018-28194-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 06/19/2018] [Indexed: 12/12/2022] Open
Abstract
Methods that provide controlled influx of molecules into cells are of critical importance for uncovering cellular mechanisms, drug development and synthetic biology. However, reliable intracellular delivery without adversely affecting the cells is a major challenge. We developed a platform for on-demand intracellular delivery applications, with which cell membrane penetration is achieved by inductive heating of micro needles. The micro needles of around 1 μm in diameter and 5 μm in length are made of gold using a silicon-based micro fabrication process that provides flexibility with respect to the needles' dimensions, pitch, shell thickness and the covered area. Experiments with HCT 116 colon cancer cells showed a high biocompatibility of the gold needle platform. Transmission electron microscopy of the cell-needle interface revealed folding of the cell membrane around the needle without penetration, preventing any delivery, which was confirmed using the EthD-1 fluorescent dye. The application of an alternating magnetic field, however, resulted in the delivery of EthD-1 by localized heating of the micro needles. Fluorescence quantification showed that intracellular delivery, with as high as 75% efficiency, is achieved for specific treatment times between 1 and 5 minutes. Overexposure of the cells to the heated micro needles, i.e. longer magnetic field application, leads to an increase in cell death, which can be exploited for cleaning the platform. This method allows to perform intracellular deliver by remotely activating the micro needles via a magnetic field, and it is controlled by the application time, making it a versatile and easy to use method. The wireless actuation could also be an attractive feature for in-vivo delivery and implantable devices.
Collapse
Affiliation(s)
- Mincho N Kavaldzhiev
- Computer, Electrical and Mathematical Science and Engineering, King Abdullah University of Science and Technology, Thuwal, 23955-6900, Saudi Arabia
| | - Jose E Perez
- Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal, 23955-6900, Saudi Arabia
| | - Rachid Sougrat
- Imaging and Characterization Core Lab-EM, King Abdullah University of Science and Technology, Thuwal, 23955-6900, Saudi Arabia
| | - Ptissam Bergam
- Imaging and Characterization Core Lab-EM, King Abdullah University of Science and Technology, Thuwal, 23955-6900, Saudi Arabia
| | - Timothy Ravasi
- Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal, 23955-6900, Saudi Arabia
| | - Jürgen Kosel
- Computer, Electrical and Mathematical Science and Engineering, King Abdullah University of Science and Technology, Thuwal, 23955-6900, Saudi Arabia.
| |
Collapse
|
48
|
McGuire AF, Santoro F, Cui B. Interfacing Cells with Vertical Nanoscale Devices: Applications and Characterization. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2018; 11:101-126. [PMID: 29570360 PMCID: PMC6530470 DOI: 10.1146/annurev-anchem-061417-125705] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Measurements of the intracellular state of mammalian cells often require probes or molecules to breach the tightly regulated cell membrane. Mammalian cells have been shown to grow well on vertical nanoscale structures in vitro, going out of their way to reach and tightly wrap the structures. A great deal of research has taken advantage of this interaction to bring probes close to the interface or deliver molecules with increased efficiency or ease. In turn, techniques have been developed to characterize this interface. Here, we endeavor to survey this research with an emphasis on the interface as driven by cellular mechanisms.
Collapse
Affiliation(s)
- Allister F McGuire
- Department of Chemistry, Stanford University, Stanford, California 94305, USA;
| | - Francesca Santoro
- Department of Chemistry, Stanford University, Stanford, California 94305, USA;
- Center for Advanced Biomaterials for Healthcare, Istituto Italiano di Tecnologia, 80125 Naples, Italy;
| | - Bianxiao Cui
- Department of Chemistry, Stanford University, Stanford, California 94305, USA;
| |
Collapse
|