1
|
Blanco-Formoso M, Galluzzi F, Vacca F, Gianiorio T, Piergentili I, Cook AB, Welzen PLW, van Hest JCM, Di Marco S, Tantussi F, Benfenati F, Colombo E, De Angelis F. Spiropyran-based glutamate nanovalve for neuronal stimulation. MATERIALS HORIZONS 2025. [PMID: 40314589 DOI: 10.1039/d5mh00082c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
One of the main challenges in medical applications is achieving precise spatial and temporal control over the release of active molecules, such as neurotransmitters. To address this issue, we engineered a nanovalve that can deliver active molecules on demand by activating or deactivating a light-sensitive chemical barrier. This valve is composed of a polymer containing a spiropyran moiety, which can switch from a hydrophobic to a hydrophilic state upon photo-stimulation. Accordingly, the nanovalve either blocks or allows molecular diffusion through a solid-state nanopore array. Here, we demonstrate that the system blocks up to 96% of the translocation of the neurotransmitter glutamate and that the on-demand release of glutamate upon light stimulation reaches 60 μM h-1, mimicking a physiological synaptic release rate. We proved its cytocompatibility and analyzed its potential for the stimulation of primary neurons and blind retinal explants by patch-clamp experiments. These results represent a milestone for the development of biomimetic neuroprostheses restoring chemical synaptic transmission lost by degeneration or delivering drugs in a light-controlled fashion.
Collapse
Affiliation(s)
- M Blanco-Formoso
- Plasmon Nanotechnology, Istituto Italiano di Tecnologia, Genova, Italy.
- CINBIO Universidade de Vigo, Vigo, Spain
| | - F Galluzzi
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.
- The Open University Affiliated Research Centre at Istituto Italiano di Tecnologia (ARC@IIT), Genova, Italy
| | - F Vacca
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - T Gianiorio
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.
- Department of Neuroscience (DINOGMI), University of Genoa, Genova, Italy
| | - I Piergentili
- Bio-Organic Chemistry, Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, The Netherlands
| | - A B Cook
- Bio-Organic Chemistry, Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, The Netherlands
| | - P L W Welzen
- Bio-Organic Chemistry, Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, The Netherlands
| | - J C M van Hest
- Bio-Organic Chemistry, Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, The Netherlands
| | - S Di Marco
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - F Tantussi
- Plasmon Nanotechnology, Istituto Italiano di Tecnologia, Genova, Italy.
| | - F Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - E Colombo
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - F De Angelis
- Plasmon Nanotechnology, Istituto Italiano di Tecnologia, Genova, Italy.
| |
Collapse
|
2
|
Vacca F, Galluzzi F, Blanco-Formoso M, Gianiorio T, De Fazio A, Tantussi F, Stürmer S, Haq W, Zrenner E, Chaffiol A, Joffrois C, Picaud S, Benfenati F, De Angelis F, Colombo E. Solid-State Nanopores for Spatially Resolved Chemical Neuromodulation. NANO LETTERS 2024; 24:15215-15225. [PMID: 39561980 PMCID: PMC11622382 DOI: 10.1021/acs.nanolett.4c02604] [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: 06/03/2024] [Revised: 10/29/2024] [Accepted: 11/04/2024] [Indexed: 11/21/2024]
Abstract
Most neural prosthetic devices are based on electrical stimulation, although the modulation of neuronal activity by a localized chemical delivery would better mimic physiological synaptic machinery. In the past decade, various drug delivery approaches attempted to emulate synaptic transmission, although they were hampered by poor retention of their cargo while reaching the target destination, low spatial resolution, and poor biocompatibility and stability of the materials involved. Here, we propose a planar solid-state device for multisite neurotransmitter translocation at the nanoscale consisting of a nanopatterned ceramic membrane connected to a reservoir designed to store neurotransmitters. We achieved diffusion-mediated glutamate stimulation of primary neurons, while we showed the feasibility to translocate other molecules through the pores by either pressure or diffusion, proving the versatility of the proposed technology. Finally, the system proved to be a promising neuronal stimulation interface in mice and nonhuman primates ex vivo, paving the way toward a biomimetic chemical stimulation in neural prosthetics and brain machine interfaces.
Collapse
Affiliation(s)
- F. Vacca
- Center for
Synaptic Neuroscience and Technology, Istituto
Italiano di Tecnologia, 16132 Genova, Italy
- IRCCS
Ospedale
Policlinico San Martino, 16132 Genova, Italy
| | - F. Galluzzi
- Center for
Synaptic Neuroscience and Technology, Istituto
Italiano di Tecnologia, 16132 Genova, Italy
- The
Open
University Affiliated Research Centre at Istituto Italiano di Tecnologia
(ARC@IIT), 16132 Genova, Italy
| | - M. Blanco-Formoso
- Plasmon Nanotechnology, Istituto Italiano di Tecnologia, 16163 Genova, Italy
- CINBIO Universidade
de Vigo, 36310 Vigo, Spain
| | - T. Gianiorio
- Center for
Synaptic Neuroscience and Technology, Istituto
Italiano di Tecnologia, 16132 Genova, Italy
- Department
of Neuroscience (DINOGMI), University of
Genoa, 16132 Genova, Italy
| | - A.F. De Fazio
- Plasmon Nanotechnology, Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - F. Tantussi
- Plasmon Nanotechnology, Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - S. Stürmer
- Centre for
Ophthalmology, Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany
| | - W. Haq
- Centre for
Ophthalmology, Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany
| | - E. Zrenner
- Centre for
Ophthalmology, Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany
| | - A. Chaffiol
- Institut
de la Vision, Sorbonne Université, 75012 Paris, France
| | - C. Joffrois
- Institut
de la Vision, Sorbonne Université, 75012 Paris, France
| | - S. Picaud
- Institut
de la Vision, Sorbonne Université, 75012 Paris, France
| | - F. Benfenati
- Center for
Synaptic Neuroscience and Technology, Istituto
Italiano di Tecnologia, 16132 Genova, Italy
- IRCCS
Ospedale
Policlinico San Martino, 16132 Genova, Italy
| | - F. De Angelis
- Plasmon Nanotechnology, Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - E. Colombo
- Center for
Synaptic Neuroscience and Technology, Istituto
Italiano di Tecnologia, 16132 Genova, Italy
- IRCCS
Ospedale
Policlinico San Martino, 16132 Genova, Italy
| |
Collapse
|
3
|
Ekstrand F, Mapar M, Ruhrmann S, Bacos K, Ling C, Prinz CN. Achieving efficient clonal beta cells transfection using nanostraw/nanopore-assisted electroporation. RSC Adv 2024; 14:22244-22252. [PMID: 39010923 PMCID: PMC11247384 DOI: 10.1039/d4ra02791d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 07/08/2024] [Indexed: 07/17/2024] Open
Abstract
The prospect of being able to efficiently inject large plasmids in insulin-producing beta cells is very attractive for diabetes research. However, conventional transfection methods suffer from high cytotoxicity or low transfection efficiency, which negatively affect their outcome. In contrast, nanostraw electroporation is a gentle method that can provide a high transfection efficiency while maintaining high cell viability. While nanostraw electroporation has gone through some method optimization in the past, such as tuning the pulse frequency, amplitude, and duration, the effect of other parameters has not been thoroughly investigated. Here, we demonstrate efficient transfection of clonal beta cells and investigate the effect of voltage at a fixed inter-electrode distance, cell density, and cargo solution conductivity on transfection efficiency. We used GFP-encoding DNA plasmids stained with an intercalating dye to enable immediate analysis and assessment of the electrophoretic transport of cargo. Moreover, we ran simulations to assess how cargo buffer conductivity impacts the transfection efficiency by affecting the voltage drop on the nanostraws and cell membrane during electroporation. Both experiments and simulations show that MilliQ water as the cargo buffer yields the best transfection efficiency. We also show that the cell density should be adjusted to maximize the number of cells interfacing the nanostraws and avoid cell stacking. Finally, we compared the transfection efficiency when using nanostraws and nanopores. Whereas the amount of GFP plasmids injected using nanostraws is larger than for nanopores, the outcome in terms of GFP fluorescence 48 h after transfection was worse than for nanopores. Moreover, when using nanostraws, fewer cells were found on the substrate 48 h after transfection compared to when using nanopores. This suggests that injecting substantial amounts of plasmids in cells can affect their proliferation and/or viability, and that nanopore electroporation, as a simpler method, is an interesting alternative to nanostraws in achieving efficient and gentle clonal beta cell transfection.
Collapse
Affiliation(s)
- Frida Ekstrand
- Division of Solid State Physics, NanoLund, Lund University 221 00 Lund Sweden
| | - Mokhtar Mapar
- Division of Solid State Physics, NanoLund, Lund University 221 00 Lund Sweden
| | - Sabrina Ruhrmann
- Epigenetics and Diabetes Unit, Lund University Diabetes Centre, Department of Clinical Sciences, Scania University Hospital 214 28 Malmö Sweden
| | - Karl Bacos
- Epigenetics and Diabetes Unit, Lund University Diabetes Centre, Department of Clinical Sciences, Scania University Hospital 214 28 Malmö Sweden
| | - Charlotte Ling
- Epigenetics and Diabetes Unit, Lund University Diabetes Centre, Department of Clinical Sciences, Scania University Hospital 214 28 Malmö Sweden
| | - Christelle N Prinz
- Division of Solid State Physics, NanoLund, Lund University 221 00 Lund Sweden
| |
Collapse
|
4
|
Zheng J, Fang J, Xu D, Liu H, Wei X, Qin C, Xue J, Gao Z, Hu N. Micronano Synergetic Three-Dimensional Bioelectronics: A Revolutionary Breakthrough Platform for Cardiac Electrophysiology. ACS NANO 2024; 18:15332-15357. [PMID: 38837178 DOI: 10.1021/acsnano.4c00052] [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: 06/06/2024]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of mortality and therefore pose a significant threat to human health. Cardiac electrophysiology plays a crucial role in the investigation and treatment of CVDs, including arrhythmia. The long-term and accurate detection of electrophysiological activity in cardiomyocytes is essential for advancing cardiology and pharmacology. Regarding the electrophysiological study of cardiac cells, many micronano bioelectric devices and systems have been developed. Such bioelectronic devices possess unique geometric structures of electrodes that enhance quality of electrophysiological signal recording. Though planar multielectrode/multitransistors are widely used for simultaneous multichannel measurement of cell electrophysiological signals, their use for extracellular electrophysiological recording exhibits low signal strength and quality. However, the integration of three-dimensional (3D) multielectrode/multitransistor arrays that use advanced penetration strategies can achieve high-quality intracellular signal recording. This review provides an overview of the manufacturing, geometric structure, and penetration paradigms of 3D micronano devices, as well as their applications for precise drug screening and biomimetic disease modeling. Furthermore, this review also summarizes the current challenges and outlines future directions for the preparation and application of micronano bioelectronic devices, with an aim to promote the development of intracellular electrophysiological platforms and thereby meet the demands of emerging clinical applications.
Collapse
Affiliation(s)
- Jilin Zheng
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou 310058, China
| | - Jiaru Fang
- School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou 510006, China
| | - Dongxin Xu
- School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou 510006, China
| | - Haitao Liu
- General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou 310052, China
| | - Xinwei Wei
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chunlian Qin
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou 310058, China
- General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou 310052, China
| | - Jiajin Xue
- General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou 310052, China
| | - Zhigang Gao
- General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou 310052, China
| | - Ning Hu
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou 310058, China
- General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou 310052, China
| |
Collapse
|
5
|
Chen Y, Shokouhi AR, Voelcker NH, Elnathan R. Nanoinjection: A Platform for Innovation in Ex Vivo Cell Engineering. Acc Chem Res 2024; 57:1722-1735. [PMID: 38819691 PMCID: PMC11191407 DOI: 10.1021/acs.accounts.4c00190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 06/01/2024]
Abstract
In human cells, intracellular access and therapeutic cargo transport, including gene-editing tools (e.g., CRISPR-Cas9 and transposons), nucleic acids (e.g., DNA, mRNA, and siRNA), peptides, and proteins (e.g., enzymes and antibodies), are tightly constrained to ensure healthy cell function and behavior. This principle is exemplified in the delivery mechanisms of chimeric antigen receptor (CAR)-T cells for ex-vivo immunotherapy. In particular, the clinical success of CAR-T cells has established a new standard of care by curing previously incurable blood cancers. The approach involves the delivery, typically via the use of electroporation (EP) and lentivirus, of therapeutic CAR genes into a patient's own T cells, which are then engineered to express CARs that target and combat their blood cancer. But the key difficulty lies in genetically manipulating these cells without causing irreversible damage or loss of function─all the while minimizing complexities of manufacturing, safety concerns, and costs, and ensuring the efficacy of the final CAR-T cell product.Nanoinjection─the process of intracellular delivery using nanoneedles (NNs)─is an emerging physical delivery route that efficiently negotiates the plasma membrane of many cell types, including primary human T cells. It occurs with minimal perturbation, invasiveness, and toxicity, with high efficiency and throughput at high spatial and temporal resolutions. Nanoinjection promises greatly improved delivery of a broad range of therapeutic cargos with little or no damage to those cargos. A nanoinjection platform allows these cargos to function in the intracellular space as desired. The adaptability of nanoinjection platforms is now bringing major advantages in immunomodulation, mechanotransduction, sampling of cell states (nanobiopsy), controlled intracellular interrogation, and the primary focus of this account─intracellular delivery and its applications in ex vivo cell engineering. Mechanical nanoinjection typically exerts direct mechanical force on the cell membrane, offering a straightforward route to improve membrane perturbation by the NNs and subsequent transport of genetic cargo into targeted cell type (adherent or suspension cells). By contrast, electroactive nanoinjection is controlled by coupling NNs with an electric field─a new route for activating electroporation (EP) at the nanoscale─allowing a dramatic reduction of the applied voltage to a cell and so minimizing post-EP damage to cells and cargo, and overcoming many of the limitations of conventional bulk EP. Nanoinjection transcends mere technique; it is an approach to cell engineering ex vivo, offering the potential to endow cells with new, powerful features such as generating chimeric antigen receptor (CAR)-T cells for future CAR-T cell technologies. We first discuss the manufacturing of NN devices (Section 2), then delve into nanoinjection-mediated cell engineering (Section 3), nanoinjection mechanisms and interfacing methodologies (Section 4), and emerging applications in using nanoinjection to create functional CAR-T cells (Section 5).
Collapse
Affiliation(s)
- Yaping Chen
- Oujiang
Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain
Health), Institute of Aging, Key Laboratory of Alzheimer’s
Disease of Zhejiang Province, Zhejiang Provincial Clinical Research
Center for Mental Disorders, The Affiliated Wenzhou Kangning Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
- Monash
Institute of Pharmaceutical Sciences, Monash
University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Ali-Reza Shokouhi
- Monash
Institute of Pharmaceutical Sciences, Monash
University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Nicolas H. Voelcker
- Monash
Institute of Pharmaceutical Sciences, Monash
University, 381 Royal Parade, Parkville, VIC 3052, Australia
- Melbourne
Centre for Nanofabrication, Victorian Node of the Australian National
Fabrication Facility, 151 Wellington Road, Clayton, VIC 3168, Australia
- Department
of Materials Science and Engineering, Monash
University, 22 Alliance Lane, Clayton, VIC 3168, Australia
| | - Roey Elnathan
- Monash
Institute of Pharmaceutical Sciences, Monash
University, 381 Royal Parade, Parkville, VIC 3052, Australia
- Melbourne
Centre for Nanofabrication, Victorian Node of the Australian National
Fabrication Facility, 151 Wellington Road, Clayton, VIC 3168, Australia
- School
of Medicine, Faculty of Health, Deakin University, Waurn Ponds, VIC 3216, Australia
- Institute
for Frontier Materials, Deakin University, Geelong Waurn Ponds campus, VIC 3216, Australia
- The
Institute for Mental and Physical Health and Clinical Translation,
School of Medicine, Deakin University, Geelong Waurn Ponds Campus, Melbourne, VIC 3216, Australia
| |
Collapse
|
6
|
Zhu X, Shi Z, Mao Y, Lächelt U, Huang R. Cell Membrane Perforation: Patterns, Mechanisms and Functions. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310605. [PMID: 38344881 DOI: 10.1002/smll.202310605] [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: 11/18/2023] [Revised: 12/21/2023] [Indexed: 02/21/2024]
Abstract
Cell membrane is crucial for the cellular activities, and any disruption to it may affect the cells. It is demonstrated that cell membrane perforation is associated with some biological processes like programmed cell death (PCD) and infection of pathogens. Specific developments make it a promising technique to perforate the cell membrane controllably and precisely. The pores on the cell membrane provide direct pathways for the entry and exit of substances, and can also cause cell death, which means reasonable utilization of cell membrane perforation is able to assist intracellular delivery, eliminate diseased or cancerous cells, and bring about other benefits. This review classifies the patterns of cell membrane perforation based on the mechanisms into 1) physical patterns, 2) biological patterns, and 3) chemical patterns, introduces the characterization methods and then summarizes the functions according to the characteristics of reversible and irreversible pores, with the aim of providing a comprehensive summary of the knowledge related to cell membrane perforation and enlightening broad applications in biomedical science.
Collapse
Affiliation(s)
- Xinran Zhu
- Key Laboratory of Smart Drug Delivery (Ministry of Education), Huashan Hospital, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Zhifeng Shi
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 201203, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 201203, China
| | - Ulrich Lächelt
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, 1090, Austria
| | - Rongqin Huang
- Key Laboratory of Smart Drug Delivery (Ministry of Education), Huashan Hospital, School of Pharmacy, Fudan University, Shanghai, 201203, China
| |
Collapse
|
7
|
Chau CC, Maffeo CM, Aksimentiev A, Radford SE, Hewitt EW, Actis P. Single molecule delivery into living cells. Nat Commun 2024; 15:4403. [PMID: 38782907 PMCID: PMC11116494 DOI: 10.1038/s41467-024-48608-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 05/02/2024] [Indexed: 05/25/2024] Open
Abstract
Controlled manipulation of cultured cells by delivery of exogenous macromolecules is a cornerstone of experimental biology. Here we describe a platform that uses nanopipettes to deliver defined numbers of macromolecules into cultured cell lines and primary cells at single molecule resolution. In the nanoinjection platform, the nanopipette is used as both a scanning ion conductance microscope (SICM) probe and an injection probe. The SICM is used to position the nanopipette above the cell surface before the nanopipette is inserted into the cell into a defined location and to a predefined depth. We demonstrate that the nanoinjection platform enables the quantitative delivery of DNA, globular proteins, and protein fibrils into cells with single molecule resolution and that delivery results in a phenotypic change in the cell that depends on the identity of the molecules introduced. Using experiments and computational modeling, we also show that macromolecular crowding in the cell increases the signal-to-noise ratio for the detection of translocation events, thus the cell itself enhances the detection of the molecules delivered.
Collapse
Affiliation(s)
- Chalmers C Chau
- School of Molecular and Cellular Biology and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
- School of Electronic and Electrical Engineering and Pollard Institute, University of Leeds, Leeds, LS2 9JT, UK
- Bragg Centre for Materials Research, University of Leeds, Leeds, UK
| | - Christopher M Maffeo
- Department of Physics, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Aleksei Aksimentiev
- Department of Physics, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Sheena E Radford
- School of Molecular and Cellular Biology and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Eric W Hewitt
- School of Molecular and Cellular Biology and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK.
| | - Paolo Actis
- School of Electronic and Electrical Engineering and Pollard Institute, University of Leeds, Leeds, LS2 9JT, UK.
- Bragg Centre for Materials Research, University of Leeds, Leeds, UK.
| |
Collapse
|
8
|
Hu H, Tian Y, Chen P, Chu W. Perspective on Tailored Nanostructure-Dominated SPP Effects for SERS. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2303001. [PMID: 38031315 DOI: 10.1002/adma.202303001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 08/14/2023] [Indexed: 12/01/2023]
Abstract
Localized surface plasmon resonance (LSPR) excited by an incident light can normally produce strong surface-enhanced Raman scattering (SERS) at the nanogaps among plasmonic nano-objects (so-called hot spots), which is extensively explored. In contrast, surface plasmon polaritons (SPPs) that can be generated by an incident beam via particular structures with a conservation of wave vectors can excite SERS effects as well. SPPs actually play an indispensable role in high-performance SERS devices but receive much less attention. In this perspective, SPPs and their couplings with LSPR for SERS excitations with differing effectiveness through particular plasmonic/dielectric structures/configurations, along with relevant fabrication approaches, are profoundly reviewed and commented on from a unique perspective from in situ to ex situ excitations of SERS enabled by spatiotemporally separated multiple processes of SPPs. Quantitative design of particular configurations/architectures enabling highly efficient and effective multiple processes of SPPs is particularly emphasized as one giant leap toward ultimate full quantitative design of intrinsically high-performance SERS chips and very critical for their batch manufacturability and applications as well. The viewpoints and prospects about innovative SERS devices based on tailored structure-dominated SPPs effects and their coupling with LSPR are presented and discussed.
Collapse
Affiliation(s)
- Haifeng Hu
- Nanofabrication Laboratory, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Yi Tian
- Nanofabrication Laboratory, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Peipei Chen
- Nanofabrication Laboratory, National Center for Nanoscience and Technology, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Weiguo Chu
- Nanofabrication Laboratory, National Center for Nanoscience and Technology, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
9
|
Xiang Y, Shi K, Li Y, Xue J, Tong Z, Li H, Li Z, Teng C, Fang J, Hu N. Active Micro-Nano-Collaborative Bioelectronic Device for Advanced Electrophysiological Recording. NANO-MICRO LETTERS 2024; 16:132. [PMID: 38411852 PMCID: PMC10899154 DOI: 10.1007/s40820-024-01336-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 12/28/2023] [Indexed: 02/28/2024]
Abstract
The development of precise and sensitive electrophysiological recording platforms holds the utmost importance for research in the fields of cardiology and neuroscience. In recent years, active micro/nano-bioelectronic devices have undergone significant advancements, thereby facilitating the study of electrophysiology. The distinctive configuration and exceptional functionality of these active micro-nano-collaborative bioelectronic devices offer the potential for the recording of high-fidelity action potential signals on a large scale. In this paper, we review three-dimensional active nano-transistors and planar active micro-transistors in terms of their applications in electro-excitable cells, focusing on the evaluation of the effects of active micro/nano-bioelectronic devices on electrophysiological signals. Looking forward to the possibilities, challenges, and wide prospects of active micro-nano-devices, we expect to advance their progress to satisfy the demands of theoretical investigations and medical implementations within the domains of cardiology and neuroscience research.
Collapse
Affiliation(s)
- Yuting Xiang
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 310058, People's Republic of China
- Department of Obstetrics and Gynecology, The Tenth Affiliated Hospital, Southern Medical University, Dongguan, 523059, People's Republic of China
- Dongguan Key Laboratory of Major Diseases in Obstetrics and Gynecology, Dongguan, 523059, People's Republic of China
| | - Keda Shi
- Department of Lung Transplantation and General Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China
| | - Ying Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People's Republic of China
| | - Jiajin Xue
- General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou, 310052, People's Republic of China
| | - Zhicheng Tong
- Department of Orthopedics, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322005, People's Republic of China
| | - Huiming Li
- Department of Orthopedics, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322005, People's Republic of China
| | - Zhongjun Li
- Department of Obstetrics and Gynecology, The Tenth Affiliated Hospital, Southern Medical University, Dongguan, 523059, People's Republic of China.
- Dongguan Key Laboratory of Major Diseases in Obstetrics and Gynecology, Dongguan, 523059, People's Republic of China.
| | - Chong Teng
- Department of Orthopedics, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322005, People's Republic of China.
| | - Jiaru Fang
- School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, People's Republic of China.
| | - Ning Hu
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 310058, People's Republic of China.
- General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou, 310052, People's Republic of China.
| |
Collapse
|
10
|
Wang H, Xue J, Li Y, Shi K, Fang J, Zheng J, Lyu X, Gao Z, Xu D, Hu N. Optimizing the Cell-Nanostructure Interface: Nanoconcave/Nanoconvex Device for Intracellular Recording of Cardiomyocytes. NANO LETTERS 2023; 23:11884-11891. [PMID: 38064276 DOI: 10.1021/acs.nanolett.3c03945] [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: 12/28/2023]
Abstract
Nanostructures are powerful components for the development of high-performance nanodevices. Revealing and understanding the cell-nanostructure interface are essential for improving and guiding nanodevice design for investigations of cell physiology. For intracellular electrophysiological detection, the cell-nanostructure interface significantly affects the quality of recorded intracellular action potentials and the application of nanodevices in cardiology research and pharmacological screening. Most of the current investigations of biointerfaces focus on nanovertical structures, and few involve nanoconcave structures. Here, we design both nanoconvex and nanoconcave devices to perform intracellular electrophysiological recordings. The amplitude, signal-to-noise ratio, duration, and repeatability of the recorded intracellular electrophysiological signals provide a multifaceted characterization of the cell-nanostructure interface. We demonstrate that devices based on both convex and concave nanostructures can create tight coupling, which facilitates high-quality and stable intracellular recordings and paves the way for precise electrophysiological study.
Collapse
Affiliation(s)
- Hao Wang
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou 310058, China
| | - Jiajin Xue
- General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Ying Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Keda Shi
- Department of Lung Transplantation and General Thoracic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jiaru Fang
- School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou 510006, China
| | - Jilin Zheng
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou 310058, China
| | - Xuelian Lyu
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou 310058, China
| | - Zhigang Gao
- General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Dongxin Xu
- School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou 510006, China
| | - Ning Hu
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou 310058, China
- General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
| |
Collapse
|
11
|
Shokouhi AR, Chen Y, Yoh HZ, Brenker J, Alan T, Murayama T, Suu K, Morikawa Y, Voelcker NH, Elnathan R. Engineering Efficient CAR-T Cells via Electroactive Nanoinjection. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2304122. [PMID: 37434421 DOI: 10.1002/adma.202304122] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 07/10/2023] [Accepted: 07/10/2023] [Indexed: 07/13/2023]
Abstract
Chimeric antigen receptor (CAR)-T cell therapy has emerged as a promising cell-based immunotherapy approach for treating blood disorders and cancers, but genetically engineering CAR-T cells is challenging due to primary T cells' sensitivity to conventional gene delivery approaches. The current viral-based method can typically involve significant operating costs and biosafety hurdles, while bulk electroporation (BEP) can lead to poor cell viability and functionality. Here, a non-viral electroactive nanoinjection (ENI) platform is developed to efficiently negotiate the plasma membrane of primary human T cells via vertically configured electroactive nanotubes, enabling efficient delivery (68.7%) and expression (43.3%) of CAR genes in the T cells, with minimal cellular perturbation (>90% cell viability). Compared to conventional BEP, the ENI platform achieves an almost threefold higher CAR transfection efficiency, indicated by the significantly higher reporter GFP expression (43.3% compared to 16.3%). By co-culturing with target lymphoma Raji cells, the ENI-transfected CAR-T cells' ability to effectively suppress lymphoma cell growth (86.9% cytotoxicity) is proved. Taken together, the results demonstrate the platform's remarkable capacity to generate functional and effective anti-lymphoma CAR-T cells. Given the growing potential of cell-based immunotherapies, such a platform holds great promise for ex vivo cell engineering, especially in CAR-T cell therapy.
Collapse
Affiliation(s)
- Ali-Reza Shokouhi
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
| | - Yaping Chen
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
| | - Hao Zhe Yoh
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
| | - Jason Brenker
- Dynamic Micro Devices (DMD) Lab, Department of Mechanical & Aerospace Engineering, Monash University, 17 College Walk, Clayton, VIC, 3168, Australia
| | - Tuncay Alan
- Dynamic Micro Devices (DMD) Lab, Department of Mechanical & Aerospace Engineering, Monash University, 17 College Walk, Clayton, VIC, 3168, Australia
| | - Takahide Murayama
- Institute of Semiconductor and Electronics Technologies ULVAC Inc., 1220-1 Suyama, Susono, Shizuoka, 410-1231, Japan
| | - Koukou Suu
- Institute of Semiconductor and Electronics Technologies ULVAC Inc., 1220-1 Suyama, Susono, Shizuoka, 410-1231, Japan
| | - Yasuhiro Morikawa
- Institute of Semiconductor and Electronics Technologies ULVAC Inc., 1220-1 Suyama, Susono, Shizuoka, 410-1231, Japan
| | - Nicolas H Voelcker
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
- Department of Materials Science and Engineering, Monash University, 22 Alliance Lane, Clayton, VIC, 3168, Australia
| | - Roey Elnathan
- School of Medicine, Faculty of Health, Deakin University, Waurn Ponds, VIC, 3216, Australia
- Institute for Frontier Materials, Deakin University, Geelong Waurn Ponds campus, Waurn Ponds, VIC, 3216, Australia
- The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong Waurn Ponds Campus, Melbourne, VIC, 3216, Australia
| |
Collapse
|
12
|
Shokouhi AR, Chen Y, Yoh HZ, Murayama T, Suu K, Morikawa Y, Brenker J, Alan T, Voelcker NH, Elnathan R. Electroactive nanoinjection platform for intracellular delivery and gene silencing. J Nanobiotechnology 2023; 21:273. [PMID: 37592297 PMCID: PMC10433684 DOI: 10.1186/s12951-023-02056-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 08/07/2023] [Indexed: 08/19/2023] Open
Abstract
BACKGROUND Nanoinjection-the process of intracellular delivery using vertically configured nanostructures-is a physical route that efficiently negotiates the plasma membrane, with minimal perturbation and toxicity to the cells. Nanoinjection, as a physical membrane-disruption-mediated approach, overcomes challenges associated with conventional carrier-mediated approaches such as safety issues (with viral carriers), genotoxicity, limited packaging capacity, low levels of endosomal escape, and poor versatility for cell and cargo types. Yet, despite the implementation of nanoinjection tools and their assisted analogues in diverse cellular manipulations, there are still substantial challenges in harnessing these platforms to gain access into cell interiors with much greater precision without damaging the cell's intricate structure. Here, we propose a non-viral, low-voltage, and reusable electroactive nanoinjection (ENI) platform based on vertically configured conductive nanotubes (NTs) that allows for rapid influx of targeted biomolecular cargos into the intracellular environment, and for successful gene silencing. The localization of electric fields at the tight interface between conductive NTs and the cell membrane drastically lowers the voltage required for cargo delivery into the cells, from kilovolts (for bulk electroporation) to only ≤ 10 V; this enhances the fine control over membrane disruption and mitigates the problem of high cell mortality experienced by conventional electroporation. RESULTS Through both theoretical simulations and experiments, we demonstrate the capability of the ENI platform to locally perforate GPE-86 mouse fibroblast cells and efficiently inject a diverse range of membrane-impermeable biomolecules with efficacy of 62.5% (antibody), 55.5% (mRNA), and 51.8% (plasmid DNA), with minimal impact on cells' viability post nanoscale-EP (> 90%). We also show gene silencing through the delivery of siRNA that targets TRIOBP, yielding gene knockdown efficiency of 41.3%. CONCLUSIONS We anticipate that our non-viral and low-voltage ENI platform is set to offer a new safe path to intracellular delivery with broader selection of cargo and cell types, and will open opportunities for advanced ex vivo cell engineering and gene silencing.
Collapse
Affiliation(s)
- Ali-Reza Shokouhi
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
| | - Yaping Chen
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
| | - Hao Zhe Yoh
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
| | - Takahide Murayama
- Institute of Semiconductor and Electronics Technologies, ULVAC Inc, 1220-1 Suyama, Susono, Shizuoka, 410-1231, Japan
| | - Koukou Suu
- Institute of Semiconductor and Electronics Technologies, ULVAC Inc, 1220-1 Suyama, Susono, Shizuoka, 410-1231, Japan
| | - Yasuhiro Morikawa
- Institute of Semiconductor and Electronics Technologies, ULVAC Inc, 1220-1 Suyama, Susono, Shizuoka, 410-1231, Japan
| | - Jason Brenker
- Department of Mechanical and Aerospace Engineering, Monash University, Wellington Rd, Clayton, VIC, 3168, Australia
| | - Tuncay Alan
- Department of Mechanical and Aerospace Engineering, Monash University, Wellington Rd, Clayton, VIC, 3168, Australia
| | - Nicolas H Voelcker
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia.
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia.
- INM-Leibniz Institute for New Materials, Campus D2 2, 66123, Saarbrücken, Germany.
- Department of Materials Science and Engineering, Monash University, 22 Alliance Lane, Clayton, VIC, 3168, Australia.
| | - Roey Elnathan
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia.
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia.
- Faculty of Health, School of Medicine, Deakin University, Waurn Ponds, Melbourne, VIC, 3216, Australia.
- Institute for Frontier Materials, Deakin University, Geelong Waurn Ponds campus, Melbourne, VIC, 3216, Australia.
- The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong Waurn Ponds Campus, Melbourne, VIC, 3216, Australia.
| |
Collapse
|
13
|
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
|
14
|
Moreddu R, Boschi A, d’Amora M, Hubarevich A, Dipalo M, De Angelis F. Passive Recording of Bioelectrical Signals from Non-Excitable Cells by Fluorescent Mirroring. NANO LETTERS 2023; 23:3217-3223. [PMID: 37019439 PMCID: PMC10141418 DOI: 10.1021/acs.nanolett.2c05053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Bioelectrical variations trigger different cell responses, including migration, mitosis, and mutation. At the tissue level, these actions result in phenomena such as wound healing, proliferation, and pathogenesis. Monitoring these mechanisms dynamically is highly desirable in diagnostics and drug testing. However, existing technologies are invasive: either they require physical access to the intracellular compartments, or they imply direct contact with the cellular medium. Here, we present a novel approach for the passive recording of electrical signals from non-excitable cells adhering to 3D microelectrodes, based on optical mirroring. Preliminary results yielded a fluorescence intensity output increase of the 5,8% in the presence of a HEK-293 cell on the electrode compared to bare microelectrodes. At present, this technology may be employed to evaluate cell-substrate adhesion and monitor cell proliferation. Further refinements could allow extrapolating quantitative data on surface charges and resting potential to investigate the electrical phenomena involved in cell migration and cancer progression.
Collapse
Affiliation(s)
- Rosalia Moreddu
- Plasmon
Nanotechnologies Unit, Istituto Italiano
di Tecnologia, 16163 Genoa, Italy
| | - Alessio Boschi
- Plasmon
Nanotechnologies Unit, Istituto Italiano
di Tecnologia, 16163 Genoa, Italy
- Department
of Bioengineering, University of Genoa, 16126 Genoa, Italy
| | - Marta d’Amora
- Plasmon
Nanotechnologies Unit, Istituto Italiano
di Tecnologia, 16163 Genoa, Italy
- Department
of Biology, University of Pisa, 56127 Pisa, Italy
| | | | - Michele Dipalo
- Plasmon
Nanotechnologies Unit, Istituto Italiano
di Tecnologia, 16163 Genoa, Italy
- Email
| | - Francesco De Angelis
- Plasmon
Nanotechnologies Unit, Istituto Italiano
di Tecnologia, 16163 Genoa, Italy
- Email
| |
Collapse
|
15
|
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: 2.5] [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
|
16
|
Maity A, Perotto S, Moschetta M, Hua H, Sardar S, Paternò GM, Tian J, Klein M, Adamo G, Lanzani G, Soci C. Resonant Enhancement of Polymer-Cell Optostimulation by a Plasmonic Metasurface. ACS OMEGA 2022; 7:42674-42680. [PMID: 36467911 PMCID: PMC9713778 DOI: 10.1021/acsomega.2c04812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/23/2022] [Indexed: 06/17/2023]
Abstract
Organic semiconductors have shown great potential as efficient bioelectronic materials. Specifically, photovoltaic polymers such as the workhorse poly(thiophene) derivatives, when stimulated with visible light, can depolarize neurons and generate action potentials, an effect that has been also employed for rescuing vision in blind rats. In this context, however, the coupling of such materials with optically resonant structures to enhance those photodriven biological effects is still in its infancy. Here, we employ the optical coupling between a nanostructured metasurface and poly(3-hexylthiophene) (P3HT) to improve the bioelectronic effects occurring upon photostimulation at the abiotic-biotic interface. In particular, we designed a spectrally tuned aluminum metasurface that can resonate with P3HT, hence augmenting the effective field experienced by the polymer. In turn, this leads to an 8-fold increase in invoked inward current in cells. This enhanced activation strategy could be useful to increase the effectiveness of P3HT-based prosthetic implants for degenerative retinal disorders.
Collapse
Affiliation(s)
- Arijit Maity
- Centre
for Disruptive Photonic Technologies, TPI, Nanyang Technological University, 21 Nanyang Link, 637371 Singapore
- Division
of Physics and Applied Physics, School of Physical and Mathematical
Sciences, Nanyang Technological University, 21 Nanyang Link, 637371 Singapore
| | - Sara Perotto
- Center
for Nano Science and Technology, Istituto
Italiano di Tecnologia, via Pascoli 70/3, 20133 Milan, Italy
| | - Matteo Moschetta
- Center
for Nano Science and Technology, Istituto
Italiano di Tecnologia, via Pascoli 70/3, 20133 Milan, Italy
| | - Huang Hua
- Department
of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 28 Medical Drive, 117456 Singapore
| | - Samim Sardar
- Center
for Nano Science and Technology, Istituto
Italiano di Tecnologia, via Pascoli 70/3, 20133 Milan, Italy
| | - Giuseppe Maria Paternò
- Center
for Nano Science and Technology, Istituto
Italiano di Tecnologia, via Pascoli 70/3, 20133 Milan, Italy
- Department
of Physics, Politecnico di Milano and Center for Nano Science and
Technology, Istituto Italiano di Tecnologia, 20133 Milan, Italy
| | - Jingyi Tian
- Centre
for Disruptive Photonic Technologies, TPI, Nanyang Technological University, 21 Nanyang Link, 637371 Singapore
- Division
of Physics and Applied Physics, School of Physical and Mathematical
Sciences, Nanyang Technological University, 21 Nanyang Link, 637371 Singapore
| | - Maciej Klein
- Centre
for Disruptive Photonic Technologies, TPI, Nanyang Technological University, 21 Nanyang Link, 637371 Singapore
- Division
of Physics and Applied Physics, School of Physical and Mathematical
Sciences, Nanyang Technological University, 21 Nanyang Link, 637371 Singapore
| | - Giorgio Adamo
- Centre
for Disruptive Photonic Technologies, TPI, Nanyang Technological University, 21 Nanyang Link, 637371 Singapore
- Division
of Physics and Applied Physics, School of Physical and Mathematical
Sciences, Nanyang Technological University, 21 Nanyang Link, 637371 Singapore
| | - Guglielmo Lanzani
- Center
for Nano Science and Technology, Istituto
Italiano di Tecnologia, via Pascoli 70/3, 20133 Milan, Italy
- Department
of Physics, Politecnico di Milano and Center for Nano Science and
Technology, Istituto Italiano di Tecnologia, 20133 Milan, Italy
| | - Cesare Soci
- Centre
for Disruptive Photonic Technologies, TPI, Nanyang Technological University, 21 Nanyang Link, 637371 Singapore
- Division
of Physics and Applied Physics, School of Physical and Mathematical
Sciences, Nanyang Technological University, 21 Nanyang Link, 637371 Singapore
| |
Collapse
|
17
|
Mejia E, Song J, Zhao Y, Qian Y, Xiao C, Lezec HJ, Agrawal A, Zhou W. Scalable two-tier protruding micro-/nano-optoelectrode arrays with hybrid optical-electrical modalities by hierarchical modular design. NANOSCALE 2022; 14:15373-15383. [PMID: 36218083 DOI: 10.1039/d2nr03820j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
In situ spatiotemporal characterization of correlated bioelectrical and biochemical processes in living multicellular systems remains a formidable challenge but can offer crucial opportunities in biology and medicine. A promising approach is to develop bio-interfaced multifunctional micro-/nano-sensor arrays with complementary biophotonic-bioelectronic modalities and biomimetic topology to achieve combined bioelectrical and biochemical detection and tight device-cell coupling. However, a system-level engineering strategy is still missing to create multifunctional micro-/nano-sensor arrays that meet the multifaceted design requirements for in situ spatiotemporal characterizations of living systems. Here, we demonstrate a hierarchical modular design and fabrication approach to develop scalable two-tier protruding micro-/nano-optoelectrode arrays that extend the design space of biomimetic micro-/nano-pillar topology, plasmonic nanoantenna-based biophotonic function in surface-enhanced Raman spectroscopy (SERS), and micro-/nano-electrode-based bioelectronics function in electrochemical impedance spectroscopy (EIS). Notably, two-tier protruding micro-/nano-optoelectrode arrays composed of nanolaminate nanoantenna arrays on top of micropillar electrode arrays can support plasmonic nanocavity modes with high SERS enhancement factors (≈106) and large surface-to-volume ratio with significantly reduced interfacial impedance in EIS measurements. We envision that scalable two-tier protruding micro-/nano-optoelectrode arrays can potentially serve as bio-interfaced multifunctional micro-/nano-sensor arrays for in situ correlated spatiotemporal bioelectrical-biochemical measurements of living multicellular systems such as neuronal network cultures, cancerous organoids, and microbial biofilms.
Collapse
Affiliation(s)
- Elieser Mejia
- Department of Electrical and Computer Engineering, Virginia Tech, Blacksburg, VA 24061, USA.
| | - Junyeob Song
- Department of Electrical and Computer Engineering, University of Delaware, Newark, DE 19716, USA
- National Institute of Standards and Technology, Gaithersburg, MD 20899, USA
| | - Yuming Zhao
- Department of Electrical and Computer Engineering, Virginia Tech, Blacksburg, VA 24061, USA.
| | - Yizhou Qian
- Department of Electrical and Computer Engineering, Virginia Tech, Blacksburg, VA 24061, USA.
| | - Chuan Xiao
- Department of Electrical and Computer Engineering, Virginia Tech, Blacksburg, VA 24061, USA.
| | - Henri J Lezec
- National Institute of Standards and Technology, Gaithersburg, MD 20899, USA
| | - Amit Agrawal
- National Institute of Standards and Technology, Gaithersburg, MD 20899, USA
| | - Wei Zhou
- Department of Electrical and Computer Engineering, Virginia Tech, Blacksburg, VA 24061, USA.
| |
Collapse
|
18
|
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
|
19
|
Chen N, He Y, Zang M, Zhang Y, Lu H, Zhao Q, Wang S, Gao Y. Approaches and materials for endocytosis-independent intracellular delivery of proteins. Biomaterials 2022; 286:121567. [DOI: 10.1016/j.biomaterials.2022.121567] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 04/26/2022] [Accepted: 05/03/2022] [Indexed: 12/12/2022]
|
20
|
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: 0.7] [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
|
21
|
Self-Assembly of Au–Ag Alloy Hollow Nanochains for Enhanced Plasmon-Driven Surface-Enhanced Raman Scattering. NANOMATERIALS 2022; 12:nano12081244. [PMID: 35457952 PMCID: PMC9025440 DOI: 10.3390/nano12081244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/01/2022] [Accepted: 04/05/2022] [Indexed: 11/17/2022]
Abstract
In this paper, Au–Ag alloy hollow nanochains (HNCs) were successfully prepared by a template-free self-assembly method achieved by partial substitution of ligands. The obtained Au–Ag alloy HNCs exhibit stronger enhancement as surface-enhanced Raman scattering (SERS) substrates than Au–Ag alloy hollow nanoparticles (HNPs) and Au nanochains substrates with an intensity ratio of about 1.3:1:1. Finite difference time domain (FDTD) simulations show that the SERS enhancement of Au–Ag alloy HNCs substrates is produced by a synergistic effect between the plasmon hybridization effect associated with the unique alloy hollow structure and the strong “hot spot” in the interstitial regions of the nanochains.
Collapse
|
22
|
Han M, Evans S, Mustafa S, Wiederman S, Ebendorff-Heidepriem H. Controlled delivery of quantum dots using microelectrophoresis technique: Intracellular behavior and preservation of cell viability. Bioelectrochemistry 2022; 144:108035. [PMID: 34906817 DOI: 10.1016/j.bioelechem.2021.108035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 11/24/2021] [Accepted: 12/01/2021] [Indexed: 11/22/2022]
Abstract
The use of synthetic nanomaterials as contrast agents, sensors, and drug delivery vehicles in biological research primarily requires effective approaches for intracellular delivery. Recently, the well-accepted microelectrophoresis technique has been reported to exhibit the ability to deliver nanomaterials, quantum dots (QDs) as an example, into live cells, but information about cell viability and intracellular fate of delivered nanomaterials is yet to be provided. Here we show that cell viability following microelectrophoresis of QDs is strongly correlated with the amount of delivered QDs, which can be finely controlled by tuning the ejection duration to maintain long-term cell survival. We reveal that microelectrophoretic delivered QDs distribute homogeneously and present pure Brownian diffusion inside the cytoplasm without endosomal entrapment, having great potential for the study of dynamic intracellular events. We validate that microelectrophoresis is a powerful technique for the effective intracellular delivery of QDs and potentially various functional nanomaterials in biological research.
Collapse
Affiliation(s)
- Mengke Han
- Institute for Photonics and Advanced Sensing (IPAS) and School of Physical Sciences, The University of Adelaide, Adelaide, South Australia 5005, Australia; ARC Centre of Excellence for Nanoscale BioPhotonics (CNBP), The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Samuel Evans
- ARC Centre of Excellence for Nanoscale BioPhotonics (CNBP), The University of Adelaide, Adelaide, South Australia 5005, Australia; Adelaide Medical School, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Sanam Mustafa
- ARC Centre of Excellence for Nanoscale BioPhotonics (CNBP), The University of Adelaide, Adelaide, South Australia 5005, Australia; Adelaide Medical School, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Steven Wiederman
- ARC Centre of Excellence for Nanoscale BioPhotonics (CNBP), The University of Adelaide, Adelaide, South Australia 5005, Australia; Adelaide Medical School, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Heike Ebendorff-Heidepriem
- Institute for Photonics and Advanced Sensing (IPAS) and School of Physical Sciences, The University of Adelaide, Adelaide, South Australia 5005, Australia; ARC Centre of Excellence for Nanoscale BioPhotonics (CNBP), The University of Adelaide, Adelaide, South Australia 5005, Australia.
| |
Collapse
|
23
|
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: 31] [Impact Index Per Article: 10.3] [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
|
24
|
Chakrabarty P, Gupta P, Illath K, Kar S, Nagai M, Tseng FG, Santra TS. Microfluidic mechanoporation for cellular delivery and analysis. Mater Today Bio 2022; 13:100193. [PMID: 35005598 PMCID: PMC8718663 DOI: 10.1016/j.mtbio.2021.100193] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/13/2021] [Accepted: 12/20/2021] [Indexed: 01/08/2023] Open
Abstract
Highly efficient intracellular delivery strategies are essential for developing therapeutic, diagnostic, biological, and various biomedical applications. The recent advancement of micro/nanotechnology has focused numerous researches towards developing microfluidic device-based strategies due to the associated high throughput delivery, cost-effectiveness, robustness, and biocompatible nature. The delivery strategies can be carrier-mediated or membrane disruption-based, where membrane disruption methods find popularity due to reduced toxicity, enhanced delivery efficiency, and cell viability. Among all of the membrane disruption techniques, the mechanoporation strategies are advantageous because of no external energy source required for membrane deformation, thereby achieving high delivery efficiencies and increased cell viability into different cell types with negligible toxicity. The past two decades have consequently seen a tremendous boost in mechanoporation-based research for intracellular delivery and cellular analysis. This article provides a brief review of the most recent developments on microfluidic-based mechanoporation strategies such as microinjection, nanoneedle arrays, cell-squeezing, and hydroporation techniques with their working principle, device fabrication, cellular delivery, and analysis. Moreover, a brief discussion of the different mechanoporation strategies integrated with other delivery methods has also been provided. Finally, the advantages, limitations, and future prospects of this technique are discussed compared to other intracellular delivery techniques.
Collapse
Affiliation(s)
- Pulasta Chakrabarty
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai, India
| | - Pallavi Gupta
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai, India
| | - Kavitha Illath
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai, India
| | - Srabani Kar
- Department of Electrical Engineering, University of Cambridge, Cambridge, CB30FA, UK
| | - Moeto Nagai
- Department of Mechanical Engineering, Toyohashi University of Technology, Aichi, Japan
| | - Fan-Gang Tseng
- Department of Engineering and System Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Tuhin Subhra Santra
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai, India
| |
Collapse
|
25
|
Harberts J, Bours K, Siegmund M, Hedrich C, Glatza M, Schöler HR, Haferkamp U, Pless O, Zierold R, Blick RH. Culturing human iPSC-derived neural progenitor cells on nanowire arrays: mapping the impact of nanowire length and array pitch on proliferation, viability, and membrane deformation. NANOSCALE 2021; 13:20052-20066. [PMID: 34842880 DOI: 10.1039/d1nr04352h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Nanowire arrays used as cell culture substrates build a potent tool for advanced biological applications such as cargo delivery and biosensing. The unique topography of nanowire arrays, however, renders them a challenging growth environment for cells and explains why only basic cell lines have been employed in existing studies. Here, we present the culturing of human induced pluripotent stem cell-derived neural progenitor cells on rectangularly arranged nanowire arrays: In detail, we mapped the impact on proliferation, viability, and topography-induced membrane deformation across a multitude of array pitches (1, 3, 5, 10 μm) and nanowire lengths (1.5, 3, 5 μm). Against the intuitive expectation, a reduced proliferation was found on the arrays with the smallest array pitch of 1 μm and long NWs. Typically, cells settle in a fakir-like state on such densely-spaced nanowires and thus experience no substantial stress caused by nanowires indenting the cell membrane. However, imaging of F-actin showed a distinct reorganization of the cytoskeleton along the nanowire tips in the case of small array pitches interfering with regular proliferation. For larger pitches, the cell numbers depend on the NW lengths but proliferation generally continued although heavy deformations of the cell membrane were observed caused by the encapsulation of the nanowires. Moreover, we noticed a strong interaction of the nanowires with the nucleus in terms of squeezing and indenting. Remarkably, the cell viability is maintained at about 85% despite the massive deformation of the cells. Considering the enormous potential of human induced stem cells to study neurodegenerative diseases and the high cellular viability combined with a strong interaction with nanowire arrays, we believe that our results pave the way to apply nanowire arrays to human stem cells for future applications in stem cell research and regenerative medicine.
Collapse
Affiliation(s)
- Jann Harberts
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany.
| | - Katja Bours
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany.
| | - Malte Siegmund
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany.
| | - Carina Hedrich
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany.
| | - Michael Glatza
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Hans R Schöler
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Undine Haferkamp
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), ScreeningPort, Schnackenburgallee 114, 22525 Hamburg, Germany
| | - Ole Pless
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), ScreeningPort, Schnackenburgallee 114, 22525 Hamburg, Germany
| | - Robert Zierold
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany.
| | - Robert H Blick
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany.
- Material Science and Engineering, College of Engineering, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| |
Collapse
|
26
|
Pandey P, Sesena-Rubfiaro A, Khatri S, He J. Development of multifunctional nanopipettes for controlled intracellular delivery and single-entity detection. Faraday Discuss 2021; 233:315-335. [PMID: 34889345 DOI: 10.1039/d1fd00057h] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The intracellular delivery of biomolecules and nanoscale materials to individual cells has gained remarkable attention in recent years owing to its wide applications in drug delivery, clinical diagnostics, bio-imaging and single-cell analysis. It remains a challenge to control and measure the delivered amount in one cell. In this work, we developed a multifunctional nanopipette - containing both a nanopore and nanoelectrode (pyrolytic carbon) at the apex - as a facile, minimally invasive and effective platform for both controllable single-cell intracellular delivery and single-entity counting. While controlled by a micromanipulator, the baseline changes of the nanopore ionic current (I) and nanoelectrode open circuit potential (V) help to guide the nanopipette tip insertion and positioning processes. The delivery from the nanopore barrel can be facilely controlled by the applied nanopore bias. To optimize the intracellular single-entity detection during delivery, we studied the effects of the nanopipette tip geometry and solution salt concentration in controlled experiments. We have successfully delivered gold nanoparticles and biomolecules into the cell, as confirmed by the increased scattering and fluorescence signals, respectively. The delivered entities have also been detected at the single-entity level using either one or both transient I and V signals. We found that the sensitivity of the single-entity electrochemical measurement was greatly affected by the local environment of the cell and varied between cell lines.
Collapse
Affiliation(s)
- Popular Pandey
- Physics Department, Florida International University, Miami, Florida, 33199, USA.
| | | | - Santosh Khatri
- Physics Department, Florida International University, Miami, Florida, 33199, USA.
| | - Jin He
- Physics Department, Florida International University, Miami, Florida, 33199, USA. .,Biomolecular Sciences Institute, Florida International University, Miami, Florida, 33199, USA
| |
Collapse
|
27
|
Chiappini C, Chen Y, Aslanoglou S, Mariano A, Mollo V, Mu H, De Rosa E, He G, Tasciotti E, Xie X, Santoro F, Zhao W, Voelcker NH, Elnathan R. Tutorial: using nanoneedles for intracellular delivery. Nat Protoc 2021; 16:4539-4563. [PMID: 34426708 DOI: 10.1038/s41596-021-00600-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 06/30/2021] [Indexed: 02/08/2023]
Abstract
Intracellular delivery of advanced therapeutics, including biologicals and supramolecular agents, is complex because of the natural biological barriers that have evolved to protect the cell. Efficient delivery of therapeutic nucleic acids, proteins, peptides and nanoparticles is crucial for clinical adoption of emerging technologies that can benefit disease treatment through gene and cell therapy. Nanoneedles are arrays of vertical high-aspect-ratio nanostructures that can precisely manipulate complex processes at the cell interface, enabling effective intracellular delivery. This emerging technology has already enabled the development of efficient and non-destructive routes for direct access to intracellular environments and delivery of cell-impermeant payloads. However, successful implementation of this technology requires knowledge of several scientific fields, making it complex to access and adopt by researchers who are not directly involved in developing nanoneedle platforms. This presents an obstacle to the widespread adoption of nanoneedle technologies for drug delivery. This tutorial aims to equip researchers with the knowledge required to develop a nanoinjection workflow. It discusses the selection of nanoneedle devices, approaches for cargo loading and strategies for interfacing to biological systems and summarises an array of bioassays that can be used to evaluate the efficacy of intracellular delivery.
Collapse
Affiliation(s)
- Ciro Chiappini
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK.
- London Centre for Nanotechnology, King's College London, London, UK.
| | - Yaping Chen
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, Victoria, Australia
| | - Stella Aslanoglou
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, Victoria, Australia
- CSIRO Manufacturing, Clayton, Victoria, Australia
| | - Anna Mariano
- Center for Advanced Biomaterials for Healthcare, Istituto Italiano di Tecnologia, Naples, Italy
| | - Valentina Mollo
- Center for Advanced Biomaterials for Healthcare, Istituto Italiano di Tecnologia, Naples, Italy
| | - Huanwen Mu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| | - Enrica De Rosa
- Center for Musculoskeletal Regeneration, Orthopedics & Sports Medicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Gen He
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou, China
| | - Ennio Tasciotti
- IRCCS San Raffaele Pisana Hospital, Rome, Italy
- San Raffaele University, Rome, Italy
- Sclavo Pharma, Siena, Italy
| | - Xi Xie
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou, China.
| | - Francesca Santoro
- Center for Advanced Biomaterials for Healthcare, Istituto Italiano di Tecnologia, Naples, Italy.
| | - Wenting Zhao
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore.
| | - Nicolas H Voelcker
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia.
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, Victoria, Australia.
- CSIRO Manufacturing, Clayton, Victoria, Australia.
- Department of Materials Science and Engineering, Monash University, Clayton, Victoria, Australia.
| | - Roey Elnathan
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia.
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, Victoria, Australia.
- Department of Materials Science and Engineering, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
28
|
Jonker D, Jafari Z, Winczewski JP, Eyovge C, Berenschot JW, Tas NR, Gardeniers JGE, De Leon I, Susarrey-Arce A. A wafer-scale fabrication method for three-dimensional plasmonic hollow nanopillars. NANOSCALE ADVANCES 2021; 3:4926-4939. [PMID: 34485816 PMCID: PMC8386417 DOI: 10.1039/d1na00316j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 07/07/2021] [Indexed: 06/13/2023]
Abstract
Access to nanofabrication strategies for crafting three-dimensional plasmonic structures is limited. In this work, a fabrication strategy to produce 3D plasmonic hollow nanopillars (HNPs) using Talbot lithography and I-line photolithography is introduced. This method is named subtractive hybrid lithography (SHL), and permits intermixed usage of nano-and-macroscale patterns. Sputter-redeposition of gold (Au) on the SHL resist pattern yields large areas of dense periodic Au-HNPs. These Au-HNPs are arranged in a square unit cell with a 250 nm pitch. The carefully controlled fabrication process resulted in Au-HNPs with nanoscale dimensions over the Au-HNP dimensions such as an 80 ± 2 nm thick solid base with a 133 ± 4 nm diameter, and a 170 ± 10 nm high nano-rim with a 14 ± 3 nm sidewall rim-thickness. The plasmonic optical response is assessed with FDTD-modeling and reveals that the highest field enhancement is at the top of the hollow nanopillar rim. The modeled field enhancement factor (EF) is compared to the experimental analytical field enhancement factor, which shows to pair up with ca. 103 < EF < 104 and ca. 103 < EF < 105 for excitation wavelengths of 633 and 785 nm. From a broader perspective, our results can stimulate the use of Au-HNPs in the fields of plasmonic sensors and spectroscopy.
Collapse
Affiliation(s)
- D Jonker
- Mesoscale Chemical Systems, MESA+ Institute, University of Twente PO. Box 217 Enschede 7500AE The Netherlands
| | - Z Jafari
- School of Engineering and Sciences, Tecnologico de Monterrey Monterrey Nuevo Leon 64849 Mexico
| | - J P Winczewski
- Mesoscale Chemical Systems, MESA+ Institute, University of Twente PO. Box 217 Enschede 7500AE The Netherlands
| | - C Eyovge
- Mesoscale Chemical Systems, MESA+ Institute, University of Twente PO. Box 217 Enschede 7500AE The Netherlands
| | - J W Berenschot
- Mesoscale Chemical Systems, MESA+ Institute, University of Twente PO. Box 217 Enschede 7500AE The Netherlands
| | - N R Tas
- Mesoscale Chemical Systems, MESA+ Institute, University of Twente PO. Box 217 Enschede 7500AE The Netherlands
| | - J G E Gardeniers
- Mesoscale Chemical Systems, MESA+ Institute, University of Twente PO. Box 217 Enschede 7500AE The Netherlands
| | - I De Leon
- School of Engineering and Sciences, Tecnologico de Monterrey Monterrey Nuevo Leon 64849 Mexico
| | - A Susarrey-Arce
- Mesoscale Chemical Systems, MESA+ Institute, University of Twente PO. Box 217 Enschede 7500AE The Netherlands
| |
Collapse
|
29
|
Hur J, Chung AJ. Microfluidic and Nanofluidic Intracellular Delivery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2004595. [PMID: 34096197 PMCID: PMC8336510 DOI: 10.1002/advs.202004595] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 04/14/2021] [Indexed: 05/05/2023]
Abstract
Innate cell function can be artificially engineered and reprogrammed by introducing biomolecules, such as DNAs, RNAs, plasmid DNAs, proteins, or nanomaterials, into the cytosol or nucleus. This process of delivering exogenous cargos into living cells is referred to as intracellular delivery. For instance, clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 gene editing begins with internalizing Cas9 protein and guide RNA into cells, and chimeric antigen receptor-T (CAR-T) cells are prepared by delivering CAR genes into T lymphocytes for cancer immunotherapies. To deliver external biomolecules into cells, tools, including viral vectors, and electroporation have been traditionally used; however, they are suboptimal for achieving high levels of intracellular delivery while preserving cell viability, phenotype, and function. Notably, as emerging solutions, microfluidic and nanofluidic approaches have shown remarkable potential for addressing this open challenge. This review provides an overview of recent advances in microfluidic and nanofluidic intracellular delivery strategies and discusses new opportunities and challenges for clinical applications. Furthermore, key considerations for future efforts to develop microfluidics- and nanofluidics-enabled next-generation intracellular delivery platforms are outlined.
Collapse
Affiliation(s)
- Jeongsoo Hur
- School of Biomedical EngineeringKorea UniversitySeoul02841Republic of Korea
| | - Aram J. Chung
- School of Biomedical EngineeringInterdisciplinary Program in Precision Public HealthKorea UniversitySeoul02841Republic of Korea
| |
Collapse
|
30
|
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: 1.0] [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
|
31
|
Gao L, Sun J, Wang L, Fan Q, Zhu G, Guo H, Sun X. Highly sensitive real-time detection of intracellular oxidative stress and application in mycotoxin toxicity evaluation based on living single-cell electrochemical sensors. Analyst 2021; 146:1444-1454. [PMID: 33410840 DOI: 10.1039/d0an02015j] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Single-cell electrochemical sensor is widely used in the local selective detection of single living cells because of its high spatial-temporal resolution and sensitivity, as well as its ability to obtain comprehensive cellular physiological states and processes with increased accuracy. Functionalized nanoprobes can detect the oxidative stress response of cells in single-cell electrochemical sensors. Moreover, the T-2 toxin is one of the most toxic mycotoxins and widely occurs in field crops. T-2 toxin can cause mitochondrial damage in cells and increase intracellular reactive oxygen species (ROS) in various cells. As the most representative free radical of intracellular ROS, H2O2 can effectively reflect the toxic effects of intracellular T-2 toxin. In this study, a functionalized gold nanoprobe was used to dynamically monitor the production of H2O2 in a single live human hepatoma cell HepG2 stimulated by mycotoxin T-2. The concentration of H2O2 produced by HepG2 cells stimulated by T-2 toxin at 1 ppb-1 ppm was linearly correlated, R2 = 0.99055, and LOD = 0.13807 ng mL-1. Sample spiking experiments were conducted, and the recovery rate of spiking was 81.19%-130.17%. A comparative analysis of differences in the current produced by multiple toxins, HT-29 cells, as well as single cells in cell populations, was performed. This method can be applied in real-time monitoring of mycotoxin toxicity during food processing in living cells and provides a novel idea for enhancing food quality and safety in a nanoenvironment.
Collapse
Affiliation(s)
- Lu Gao
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Synergetic Innovation Center of Food Safety and Quality Control, Jiangnan University, Wuxi, Jiangsu 214122, P.R. China.
| | | | | | | | | | | | | |
Collapse
|
32
|
Ardini M, Bellelli A, Williams DL, Di Leandro L, Giansanti F, Cimini A, Ippoliti R, Angelucci F. Taking Advantage of the Morpheein Behavior of Peroxiredoxin in Bionanotechnology. Bioconjug Chem 2021; 32:43-62. [PMID: 33411522 PMCID: PMC8023583 DOI: 10.1021/acs.bioconjchem.0c00621] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
![]()
Morpheeins
are proteins that reversibly assemble into different
oligomers, whose architectures are governed by conformational changes
of the subunits. This property could be utilized in bionanotechnology
where the building of nanometric and new high-ordered structures is
required. By capitalizing on the adaptability of morpheeins to create
patterned structures and exploiting their inborn affinity toward inorganic
and living matter, “bottom-up” creation of nanostructures
could be achieved using a single protein building block, which may
be useful as such or as scaffolds for more complex materials. Peroxiredoxins
represent the paradigm of a morpheein that can be applied to bionanotechnology.
This review describes the structural and functional transitions that
peroxiredoxins undergo to form high-order oligomers, e.g., rings,
tubes, particles, and catenanes, and reports on the chemical and genetic
engineering approaches to employ them in the generation of responsive
nanostructures and nanodevices. The usefulness of the morpheeins’
behavior is emphasized, supporting their use in future applications.
Collapse
Affiliation(s)
- Matteo Ardini
- Department of Life, Health, and Environmental Sciences, University of L'Aquila, Piazzale Salvatore Tommasi 1, 67100 L'Aquila, Italy
| | - Andrea Bellelli
- Department of Biochemical Sciences "A. Rossi Fanelli", University of Roma "Sapienza", Piazzale Aldo Moro 5, 00185 Roma, Italy
| | - David L Williams
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, Illinois 60612, United States
| | - Luana Di Leandro
- Department of Life, Health, and Environmental Sciences, University of L'Aquila, Piazzale Salvatore Tommasi 1, 67100 L'Aquila, Italy
| | - Francesco Giansanti
- Department of Life, Health, and Environmental Sciences, University of L'Aquila, Piazzale Salvatore Tommasi 1, 67100 L'Aquila, Italy
| | - Annamaria Cimini
- Department of Life, Health, and Environmental Sciences, University of L'Aquila, Piazzale Salvatore Tommasi 1, 67100 L'Aquila, Italy
| | - Rodolfo Ippoliti
- Department of Life, Health, and Environmental Sciences, University of L'Aquila, Piazzale Salvatore Tommasi 1, 67100 L'Aquila, Italy
| | - Francesco Angelucci
- Department of Life, Health, and Environmental Sciences, University of L'Aquila, Piazzale Salvatore Tommasi 1, 67100 L'Aquila, Italy
| |
Collapse
|
33
|
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: 0.8] [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
|
34
|
Byrne HJ, Bonnier F, Efeoglu E, Moore C, McIntyre J. In vitro Label Free Raman Microspectroscopic Analysis to Monitor the Uptake, Fate and Impacts of Nanoparticle Based Materials. Front Bioeng Biotechnol 2020; 8:544311. [PMID: 33195114 PMCID: PMC7658377 DOI: 10.3389/fbioe.2020.544311] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 10/12/2020] [Indexed: 01/22/2023] Open
Abstract
The continued emergence of nanoscale materials for nanoparticle-based therapy, sensing and imaging, as well as their more general adoption in a broad range of industrial applications, has placed increasing demands on the ability to assess their interactions and impacts at a cellular and subcellular level, both in terms of potentially beneficial and detrimental effects. Notably, however, many such materials have been shown to interfere with conventional in vitro cellular assays that record only a single colorimetric end-point, challenging the ability to rapidly screen cytological responses. As an alternative, Raman microspectroscopy can spatially profile the biochemical content of cells, and any changes to it as a result of exogenous agents, such as toxicants or therapeutic agents, in a label free manner. In the confocal mode, analysis can be performed at a subcellular level. The technique has been employed to confirm the cellular uptake and subcellular localization of polystyrene nanoparticles (PSNPs), graphene and molybdenum disulfide micro/nano plates (MoS2), based on their respective characteristic spectroscopic signatures. In the case of PSNPs it was further employed to identify their local subcellular environment in endosomes, lysosomes and endoplasmic reticulum, while for MoS2 particles, it was employed to monitor subcellular degradation as a function of time. For amine functionalized PSNPs, the potential of Raman microspectroscopy to quantitatively characterize the dose and time dependent toxic responses has been explored, in a number of cell lines. Comparing the responses to those of poly (amidoamine) nanoscale polymeric dendrimers, differentiation of apoptotic and necrotic pathways based on the cellular spectroscopic responses was demonstrated. Drawing in particular from the experience of the authors, this paper details the progress to date in the development of applications of Raman microspectroscopy for in vitro, label free analysis of the uptake, fate and impacts of nanoparticle based materials, in vitro, and the prospects for the development of a routine, label free high content spectroscopic analysis technique.
Collapse
Affiliation(s)
- Hugh J Byrne
- FOCAS Research Institute, Technological University Dublin, Dublin, Ireland
| | - Franck Bonnier
- UFR Sciences Pharmaceutiques, EA 6295 Nanomédicaments et Nanosondes, Université de Tours, Tours, France
| | - Esen Efeoglu
- FOCAS Research Institute, Technological University Dublin, Dublin, Ireland
| | - Caroline Moore
- FOCAS Research Institute, Technological University Dublin, Dublin, Ireland
| | - Jennifer McIntyre
- FOCAS Research Institute, Technological University Dublin, Dublin, Ireland
| |
Collapse
|
35
|
Xu X, Jia J, Guo M. The Most Recent Advances in the Application of Nano-Structures/Nano-Materials for Single-Cell Sampling. Front Chem 2020; 8:718. [PMID: 32974282 PMCID: PMC7469254 DOI: 10.3389/fchem.2020.00718] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 07/13/2020] [Indexed: 11/13/2022] Open
Abstract
The research in endogenous biomolecules from a single cell has grown rapidly in recent years since it is critical for dissecting and scrutinizing the complexity of heterogeneous tissues, especially under pathological conditions, and it is also of key importance to understand the biological processes and cellular responses to various perturbations without the limitation of population averaging. Although conventional techniques, such as micromanipulation or cell sorting methods, are already used along with subsequent molecular examinations, it remains a big challenge to develop new approaches to manipulate and directly extract small quantities of cytosol from single living cells. In this sense, nanostructure or nanomaterial may play a critical role in overcoming these challenges in cellular manipulation and extraction of very small quantities of cells, and provide a powerful alternative to conventional techniques. Since the nanostructures or nanomaterial could build channels between intracellular and extracellular components across cell membrane, through which cytosol could be pumped out and transferred to downstream analyses. In this review, we will first brief the traditional methods for single cell analyses, and then shift our focus to some most promising methods for single-cell sampling with nanostructures, such as glass nanopipette, nanostraw, carbon nanotube probes and other nanomaterial. In this context, particular attentions will be paid to their principles, preparations, operations, superiorities and drawbacks, and meanwhile the great potential of nano-materials for single-cell sampling will also be highlighted and prospected.
Collapse
Affiliation(s)
- Xiaolong Xu
- School of Biotechnology and Health Science, Wuyi University, Jiangmen, China
| | - Jianbo Jia
- School of Biotechnology and Health Science, Wuyi University, Jiangmen, China
| | - Mingquan Guo
- School of Biotechnology and Health Science, Wuyi University, Jiangmen, China.,CAS Key Laboratory of Plant Germplasm Enhancement and Specialty Agriculture, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
36
|
A ring-shaped protein clusters gold nanoparticles acting as molecular scaffold for plasmonic surfaces. Biochim Biophys Acta Gen Subj 2020; 1864:129617. [DOI: 10.1016/j.bbagen.2020.129617] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 04/09/2020] [Accepted: 04/12/2020] [Indexed: 12/18/2022]
|
37
|
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: 3.0] [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
|
38
|
Liu J, Fraire JC, De Smedt SC, Xiong R, Braeckmans K. Intracellular Labeling with Extrinsic Probes: Delivery Strategies and Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2000146. [PMID: 32351015 DOI: 10.1002/smll.202000146] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/29/2020] [Accepted: 03/16/2020] [Indexed: 06/11/2023]
Abstract
Extrinsic probes have outstanding properties for intracellular labeling to visualize dynamic processes in and of living cells, both in vitro and in vivo. Since extrinsic probes are in many cases cell-impermeable, different biochemical, and physical approaches have been used to break the cell membrane barrier for direct delivery into the cytoplasm. In this Review, these intracellular delivery strategies are discussed, briefly explaining the mechanisms and how they are used for live-cell labeling applications. Methods that are discussed include three biochemical agents that are used for this purpose-purpose-different nanocarriers, cell penetrating peptides and the pore-foraming bacterial toxin streptolysin O. Most successful intracellular label delivery methods are, however, based on physical principles to permeabilize the membrane and include electroporation, laser-induced photoporation, micro- and nanoinjection, nanoneedles or nanostraws, microfluidics, and nanomachines. The strengths and weaknesses of each strategy are discussed with a systematic comparison provided. Finally, the extrinsic probes that are reported for intracellular labeling so-far are summarized, together with the delivery strategies that are used and their performance. This combined information should provide for a useful guide for choosing the most suitable delivery method for the desired probes.
Collapse
Affiliation(s)
- Jing Liu
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent, B-9000, Belgium
| | - Juan C Fraire
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent, B-9000, Belgium
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent, B-9000, Belgium
- Centre for Advanced Light Microscopy, Ghent University, Ghent, B-9000, Belgium
- Joint Laboratory of Advanced Biomedical Technology (NFU-UGent), College of Chemical Engineering, Nanjing Forestry University (NFU), Nanjing, 210037, P. R. China
| | - Ranhua Xiong
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent, B-9000, Belgium
| | - Kevin Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent, B-9000, Belgium
- Centre for Advanced Light Microscopy, Ghent University, Ghent, B-9000, Belgium
| |
Collapse
|
39
|
Chen Y, Aslanoglou S, Murayama T, Gervinskas G, Fitzgerald LI, Sriram S, Tian J, Johnston APR, Morikawa Y, Suu K, Elnathan R, Voelcker NH. Silicon-Nanotube-Mediated Intracellular Delivery Enables Ex Vivo Gene Editing. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2000036. [PMID: 32378244 DOI: 10.1002/adma.202000036] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 03/16/2020] [Accepted: 03/22/2020] [Indexed: 06/11/2023]
Abstract
Engineered nano-bio cellular interfaces driven by vertical nanostructured materials are set to spur transformative progress in modulating cellular processes and interrogations. In particular, the intracellular delivery-a core concept in fundamental and translational biomedical research-holds great promise for developing novel cell therapies based on gene modification. This study demonstrates the development of a mechanotransfection platform comprising vertically aligned silicon nanotube (VA-SiNT) arrays for ex vivo gene editing. The internal hollow structure of SiNTs allows effective loading of various biomolecule cargoes; and SiNTs mediate delivery of those cargoes into GPE86 mouse embryonic fibroblasts without compromising their viability. Focused ion beam scanning electron microscopy (FIB-SEM) and confocal microscopy results demonstrate localized membrane invaginations and accumulation of caveolin-1 at the cell-NT interface, suggesting the presence of endocytic pits. Small-molecule inhibition of endocytosis suggests that active endocytic process plays a role in the intracellular delivery of cargo from SiNTs. SiNT-mediated siRNA intracellular delivery shows the capacity to reduce expression levels of F-actin binding protein (Triobp) and alter the cellular morphology of GPE86. Finally, the successful delivery of Cas9 ribonucleoprotein (RNP) to specifically target mouse Hprt gene is achieved. This NT-enhanced molecular delivery platform has strong potential to support gene editing technologies.
Collapse
Affiliation(s)
- Yaping Chen
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, VIC, 3168, Australia
| | - Stella Aslanoglou
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, VIC, 3168, Australia
| | - Takahide Murayama
- Institute of Semiconductor and Electronics Technologies, ULVAC Inc., 1220-1 Suyama, Susono, Shizuoka, 410-1231, Japan
| | - Gediminas Gervinskas
- Ramaciotti Centre for Cryo-Electron Microscopy, Monash University, 15 Innovation Walk, Clayton, VIC, 3800, Australia
| | - Laura I Fitzgerald
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Sharath Sriram
- MicroNano Research Facility (MNRF), RMIT University, Melbourne City Campus, Building 7, Level 4, Bowen Street, Melbourne, VIC, 3000, Australia
| | - Jie Tian
- MicroNano Research Facility (MNRF), RMIT University, Melbourne City Campus, Building 7, Level 4, Bowen Street, Melbourne, VIC, 3000, Australia
| | - Angus P R Johnston
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Yasuhiro Morikawa
- 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
| | - 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
- Department of Materials Science and Engineering Monash University, 22 Alliance Lane, 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
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, VIC, 3168, Australia
- Department of Materials Science and Engineering Monash University, 22 Alliance Lane, Clayton, VIC, 3168, Australia
- INM-Leibniz Institute for New Materials, Campus D2 2, Saarbrücken, 66123, Germany
| |
Collapse
|
40
|
Methods for protein delivery into cells: from current approaches to future perspectives. Biochem Soc Trans 2020; 48:357-365. [DOI: 10.1042/bst20190039] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 03/16/2020] [Accepted: 03/18/2020] [Indexed: 12/14/2022]
Abstract
The manipulation of cultured mammalian cells by the delivery of exogenous macromolecules is one of the cornerstones of experimental cell biology. Although the transfection of cells with DNA expressions constructs that encode proteins is routine and simple to perform, the direct delivery of proteins into cells has many advantages. For example, proteins can be chemically modified, assembled into defined complexes and subject to biophysical analyses prior to their delivery into cells. Here, we review new approaches to the injection and electroporation of proteins into cultured cells. In particular, we focus on how recent developments in nanoscale injection probes and localized electroporation devices enable proteins to be delivered whilst minimizing cellular damage. Moreover, we discuss how nanopore sensing may ultimately enable the quantification of protein delivery at single-molecule resolution.
Collapse
|
41
|
Ali H, Ni H, Xu X. Near-UV luminescence tomography with an aperture-free meta super oscillatory lens for single molecule detection. JOURNAL OF THE OPTICAL SOCIETY OF AMERICA. A, OPTICS, IMAGE SCIENCE, AND VISION 2020; 37:621-628. [PMID: 32400541 DOI: 10.1364/josaa.383854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 02/22/2020] [Indexed: 06/11/2023]
Abstract
Medical physics offers super oscillatory lenses (SOL) to attain subwavelength focusing for efficient image resolution in the detection of a single molecule. In this paper, we propose a negative-indexed permeability-controlled meta SOL, which restores the light concentration via exciting surface plasmon resonance and enhances luminescence at our region of interest, i.e., fluid sample (${S_L}$SL). The meta SOL is aperture-free in comparison with other techniques (zero-mode waveguides and nano antennas) and hence avoids the constraints, which are posed by nano apertures, to insert a small volume of $ S_L $SL into these nano apertures for detection purposes. The meta SOL is a unique combination of gold split rings mounted on silicon dioxide (${{\rm SiO}_2}$SiO2) substrate and operational in the near-ultraviolet (UV) region. We utilized the phenomena of negative index of refraction, and our simulated trails exploit the magnetic (${\mu _r}$μr) response of the meta SOL by analyzing its transmission spectra in the frequency range from 3.53 to 3.57 Peta Hz. We observe the methodical response of the meta SOL with its ample potential to surpass the resolution at a working wavelength of $\lambda ={84.173}\;{\rm nm}$λ=84.173nm, which enhances luminescence by restoring the evanescent UV magnetic field (${B_{\rm UV}}$BUV) at ${S_L}$SL. This technique will offer a new and easy approach to uplift the efficiency of super oscillatory lenses in the near-UV regime to benefit single molecule detection techniques and thus the novelty.
Collapse
|
42
|
Kang G, Carlson DW, Kang TH, Lee S, Haward SJ, Choi I, Shen AQ, Chung AJ. Intracellular Nanomaterial Delivery via Spiral Hydroporation. ACS NANO 2020; 14:3048-3058. [PMID: 32069037 DOI: 10.1021/acsnano.9b07930] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
In recent nanobiotechnology developments, a wide variety of functional nanomaterials and engineered biomolecules have been created, and these have numerous applications in cell biology. For these nanomaterials to fulfill their promises completely, they must be able to reach their biological targets at the subcellular level and with a high level of specificity. Traditionally, either nanocarrier- or membrane disruption-based method has been used to deliver nanomaterials inside cells; however, these methods are suboptimal due to their toxicity, inconsistent delivery, and low throughput, and they are also labor intensive and time-consuming, highlighting the need for development of a next-generation, intracellular delivery system. This study reports on the development of an intracellular nanomaterial delivery platform, based on unexpected cell-deformation phenomena via spiral vortex and vortex breakdown exerted in the cross- and T-junctions at moderate Reynolds numbers. These vortex-induced cell deformation and sequential restoration processes open cell membranes transiently, allowing effective and robust intracellular delivery of nanomaterials in a single step without the aid of carriers or external apparatus. By using the platform described here (termed spiral hydroporator), we demonstrate the delivery of different nanomaterials, including gold nanoparticles (200 nm diameter), functional mesoporous silica nanoparticles (150 nm diameter), dextran (hydrodynamic diameters between 2-55 nm), and mRNA, into different cell types. We demonstrate here that the system is highly efficient (up to 96.5%) with high throughput (up to 1 × 106 cells/min) and rapid delivery (∼1 min) while maintaining high levels of cell viability (up to 94%).
Collapse
Affiliation(s)
- GeoumYoung Kang
- Department of Bio-convergence Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Daniel W Carlson
- Micro/Bio/Nanofluidics Unit, Okinawa Institute of Science and Technology Graduate University (OIST), Okinawa 904-0495, Japan
| | - Tae Ho Kang
- Department of Life Science, University of Seoul, Seoul 02504, Republic of Korea
| | - Seungki Lee
- Department of Life Science, University of Seoul, Seoul 02504, Republic of Korea
| | - Simon J Haward
- Micro/Bio/Nanofluidics Unit, Okinawa Institute of Science and Technology Graduate University (OIST), Okinawa 904-0495, Japan
| | - Inhee Choi
- Department of Life Science, University of Seoul, Seoul 02504, Republic of Korea
| | - Amy Q Shen
- Micro/Bio/Nanofluidics Unit, Okinawa Institute of Science and Technology Graduate University (OIST), Okinawa 904-0495, Japan
| | - Aram J Chung
- Department of Bio-convergence Engineering, Korea University, Seoul 02841, Republic of Korea
- Department of Bioengineering, Korea University, Seoul 02841, Republic of Korea
- School of Biomedical Engineering, Korea University, Seoul 02841, Republic of Korea
| |
Collapse
|
43
|
Pan R, Hu K, Jia R, Rotenberg SA, Jiang D, Mirkin MV. Resistive-Pulse Sensing Inside Single Living Cells. J Am Chem Soc 2020; 142:5778-5784. [DOI: 10.1021/jacs.9b13796] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Rongrong Pan
- Department of Chemistry and Biochemistry, Queens College-CUNY, Flushing, New York 11367, United States
- State Key Laboratory of Analytical Chemistry for Life and School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Keke Hu
- Department of Chemistry and Biochemistry, Queens College-CUNY, Flushing, New York 11367, United States
- The Graduate Center of CUNY, New York, New York 10016, United States
| | - Rui Jia
- Department of Chemistry and Biochemistry, Queens College-CUNY, Flushing, New York 11367, United States
- The Graduate Center of CUNY, New York, New York 10016, United States
| | - Susan A. Rotenberg
- Department of Chemistry and Biochemistry, Queens College-CUNY, Flushing, New York 11367, United States
- The Graduate Center of CUNY, New York, New York 10016, United States
| | - Dechen Jiang
- State Key Laboratory of Analytical Chemistry for Life and School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Michael V. Mirkin
- Department of Chemistry and Biochemistry, Queens College-CUNY, Flushing, New York 11367, United States
- The Graduate Center of CUNY, New York, New York 10016, United States
| |
Collapse
|
44
|
Higgins SG, Becce M, Belessiotis-Richards A, Seong H, Sero JE, Stevens MM. High-Aspect-Ratio Nanostructured Surfaces as Biological Metamaterials. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1903862. [PMID: 31944430 PMCID: PMC7610849 DOI: 10.1002/adma.201903862] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 10/02/2019] [Indexed: 04/14/2023]
Abstract
Materials patterned with high-aspect-ratio nanostructures have features on similar length scales to cellular components. These surfaces are an extreme topography on the cellular level and have become useful tools for perturbing and sensing the cellular environment. Motivation comes from the ability of high-aspect-ratio nanostructures to deliver cargoes into cells and tissues, access the intracellular environment, and control cell behavior. These structures directly perturb cells' ability to sense and respond to external forces, influencing cell fate, and enabling new mechanistic studies. Through careful design of their nanoscale structure, these systems act as biological metamaterials, eliciting unusual biological responses. While predominantly used to interface eukaryotic cells, there is growing interest in nonanimal and prokaryotic cell interfacing. Both experimental and theoretical studies have attempted to develop a mechanistic understanding for the observed behaviors, predominantly focusing on the cell-nanostructure interface. This review considers how high-aspect-ratio nanostructured surfaces are used to both stimulate and sense biological systems.
Collapse
Affiliation(s)
- Stuart G. Higgins
- Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK
| | | | | | - Hyejeong Seong
- Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK
| | - Julia E. Sero
- Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK
| | - Molly M. Stevens
- Department of Materials, Imperial College London, London, SW7 2AZ, UK
- Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK
- Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| |
Collapse
|
45
|
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: 8] [Impact Index Per Article: 1.6] [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
|