1
|
Liu F, Su R, Jiang X, Wang S, Mu W, Chang L. Advanced micro/nano-electroporation for gene therapy: recent advances and future outlook. NANOSCALE 2024; 16:10500-10521. [PMID: 38757536 DOI: 10.1039/d4nr01408a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
Gene therapy is a promising disease treatment approach by editing target genes, and thus plays a fundamental role in precision medicine. To ensure gene therapy efficacy, the effective delivery of therapeutic genes into specific cells is a key challenge. Electroporation utilizes short electric pulses to physically break the cell membrane barrier, allowing gene transfer into the cells. It dodges the off-target risks associated with viral vectors, and also stands out from other physical-based gene delivery methods with its high-throughput and cargo-accelerating features. In recent years, with the help of advanced micro/nanotechnology, micro/nanostructure-integrated electroporation (micro/nano-electroporation) techniques and devices have significantly improved cell viability, transfection efficiency and dose controllability of the electroporation strategy, enhancing its application practicality especially in vivo. This technical advancement makes micro/nano-electroporation an effective and versatile tool for gene therapy. In this review, we first introduce the evolution of electroporation technique with a brief explanation of the perforation mechanism, and then provide an overview of the recent advancements and prospects of micro/nano-electroporation technology in the field of gene therapy. To comprehensively showcase the latest developments of micro/nano-electroporation technology in gene therapy, we focus on discussing micro/nano-electroporation devices and current applications at both in vitro and in vivo levels. Additionally, we outline the ongoing clinical studies of gene electrotransfer (GET), revealing the tremendous potential of electroporation-based gene delivery in disease treatment and healthcare. Lastly, the challenges and future directions in this field are discussed.
Collapse
Affiliation(s)
- Feng Liu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Rongtai Su
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Xinran Jiang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Siqi Wang
- Department of General Surgery and Obesity and Metabolic Disease Center, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Wei Mu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
- School of Engineering Medicine, Beihang University, Beijing, 100191, China
- Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of the People's Republic of China, Beijing, 100191, China
| | - Lingqian Chang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| |
Collapse
|
2
|
Pan J, Wang X, Chiang CL, Ma Y, Cheng J, Bertani P, Lu W, Lee LJ. Joule heating and electroosmotic flow in cellular micro/nano electroporation. LAB ON A CHIP 2024; 24:819-831. [PMID: 38235543 DOI: 10.1039/d3lc00568b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Localized micro/nano-electroporation (MEP/NEP) shows tremendous potential in cell transfection with high cell viability, precise dose control, and good transfection efficacy. In MEP/NEP, micro or nanochannels are used to tailor the electric field distribution. Cells are positioned tightly by a micron or nanochannel, and the cargoes are delivered into the cell via the channel by electrophoresis (EP). Such confined geometries with micro and nanochannels are also widely used in sorting, isolation, and condensing of biomolecules and cells. Theoretical studies on the electrokinetic phenomena in these applications have been well established. However, for MEP/NEP applications, electrokinetic phenomena and their impact on the cell transfection efficiency and cell survival rate have not been studied comprehensively. In this work, we reveal the coupling between electric field, Joule heating, electroosmosis (EO), and EP in MEP/NEP at different channel sizes. A microfluidic biochip is used to investigate the electrokinetic phenomena in MEP/NEP on a single cell level. Bubble formation is observed at a threshold voltage due to Joule heating. The bubble is pushed to the cargo side due to EO and grows at the outlet of the nanochannel. As the voltage increases, the cargo transport efficiency decreases due to more intense EO, particularly for plasmid DNAs (3.5 kbp) with a low EP mobility. An 'electroporation zone' is defined for NEP/MEP systems with different channel sizes to avoid bubble formation and excessive EO velocity that may reduce the cargo delivery efficiency.
Collapse
Affiliation(s)
- Junjie Pan
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, USA.
| | - Xinyu Wang
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, USA.
| | - Chi-Ling Chiang
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, USA.
| | - Yifan Ma
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| | - Junao Cheng
- Department of Electrical and Computer Engineering, The Ohio State University, Columbus, Ohio 43210, USA.
| | - Paul Bertani
- Department of Electrical and Computer Engineering, The Ohio State University, Columbus, Ohio 43210, USA.
| | - Wu Lu
- Department of Electrical and Computer Engineering, The Ohio State University, Columbus, Ohio 43210, USA.
| | - L James Lee
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, USA.
| |
Collapse
|
3
|
Hinnekens C, De Smedt SC, Fraire JC, Braeckmans K. Non-viral engineering of NK cells. Biotechnol Adv 2023; 68:108212. [PMID: 37454745 DOI: 10.1016/j.biotechadv.2023.108212] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 06/06/2023] [Accepted: 07/09/2023] [Indexed: 07/18/2023]
Abstract
The last decade has witnessed great progress in the field of adoptive cell therapies, with the authorization of Kymriah (tisagenlecleucel) in 2017 by the Food and Drug Administration (FDA) as a crucial stepstone. Since then, five more CAR-T therapies have been approved for the treatment of hematological malignancies. While this is a great step forward to treating several types of blood cancers, CAR-T cell therapies are still associated with severe side-effects such as Graft-versus-Host Disease (GvHD), cytokine release syndrome (CRS) and neurotoxicity. Because of this, there has been continued interest in Natural Killer cells which avoid these side-effects while offering the possibility to generate allogeneic cell therapies. Similar to T-cells, NK cells can be genetically modified to improve their therapeutic efficacy in a variety of ways. In contrast to T cells, viral transduction of NK cells remains inefficient and induces cytotoxic effects. Viral vectors also require a lengthy and expensive product development process and are accompanied by certain risks such as insertional mutagenesis. Therefore, non-viral transfection technologies are avidly being developed aimed at addressing these shortcomings of viral vectors. In this review we will present an overview of the potential of NK cells in cancer immunotherapies and the non-viral transfection technologies that have been explored to engineer them.
Collapse
Affiliation(s)
- Charlotte Hinnekens
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Juan C Fraire
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, 08028 Barcelona, Spain.
| | - Kevin Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| |
Collapse
|
4
|
Pfisterer F, Godino N, Gerling T, Kirschbaum M. Continuous microfluidic flow-through protocol for selective and image-activated electroporation of single cells. RSC Adv 2023; 13:19379-19387. [PMID: 37383687 PMCID: PMC10294288 DOI: 10.1039/d3ra03100d] [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: 05/09/2023] [Accepted: 05/31/2023] [Indexed: 06/30/2023] Open
Abstract
Electroporation of cells is a widely-used tool to transport molecules such as proteins or nucleic acids into cells or to extract cellular material. However, bulk methods for electroporation do not offer the possibility to selectively porate subpopulations or single cells in heterogeneous cell samples. To achieve this, either presorting or complex single-cell technologies are required currently. In this work, we present a microfluidic flow protocol for selective electroporation of predefined target cells identified in real-time by high-quality microscopic image analysis of fluorescence and transmitted light. While traveling through the microchannel, the cells are focused by dielectrophoretic forces into the microscopic detection area, where they are classified based on image analysis techniques. Finally, the cells are forwarded to a poration electrode and only the target cells are pulsed. By processing a heterogenically stained cell sample, we were able to selectively porate only target cells (green-fluorescent) while non-target cells (blue-fluorescent) remained unaffected. We achieved highly selective poration with >90% specificity at average poration rates of >50% and throughputs of up to 7200 cells per hour.
Collapse
Affiliation(s)
- Felix Pfisterer
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Branch Bioanalytics and Bioprocesses IZI-BB Am Muehlenberg 13 14476 Potsdam Germany
| | - Neus Godino
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Branch Bioanalytics and Bioprocesses IZI-BB Am Muehlenberg 13 14476 Potsdam Germany
| | - Tobias Gerling
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Branch Bioanalytics and Bioprocesses IZI-BB Am Muehlenberg 13 14476 Potsdam Germany
| | - Michael Kirschbaum
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Branch Bioanalytics and Bioprocesses IZI-BB Am Muehlenberg 13 14476 Potsdam Germany
| |
Collapse
|
5
|
Lard M, Ho BD, Beech JP, Tegenfeldt JO, Prinz CN. Use of dielectrophoresis for directing T cells to microwells before nanostraw transfection: modelling and experiments. RSC Adv 2022; 12:30295-30303. [PMID: 36337971 PMCID: PMC9589401 DOI: 10.1039/d2ra05119b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/14/2022] [Indexed: 11/07/2022] Open
Abstract
Nanostraw substrates have great potential for achieving minimally invasive cell transfection. Cells located on the nanostraw substrate are subjected to mild DC electric pulses applied across the nanostraw substrate, which open pores in the cell membrane on top of the nanostraws and drives charged cargo through these pores via electrophoresis. However, with this method, the current may leak through uncovered nanostraws, thereby decreasing the desired effect in the cell-covered nanostraws. A minimization of the number of uncovered nanostraws could be achieved by high cell coverage, but this is challenging when working with small cell populations. Nanostraw substrates of smaller area could be covered by smaller cell populations but are hard to integrate into fluidics systems. Here, we use simulations and experiments to show that this issue can be addressed by covering the nanostraw substrate with an insulating layer containing pores of similar size to cells. The pores act as traps into which cells can be guided using dielectrophoresis, ensuring a high degree of occupancy while maintaining a high cell viability, even if the total number of cells is low.
Collapse
Affiliation(s)
- Mercy Lard
- Division of Solid State Physics and NanoLund, Lund University 221 00 Lund Sweden
| | - Bao D Ho
- Division of Solid State Physics and NanoLund, Lund University 221 00 Lund Sweden
| | - Jason P Beech
- Division of Solid State Physics and NanoLund, Lund University 221 00 Lund Sweden
| | - Jonas O Tegenfeldt
- Division of Solid State Physics and NanoLund, Lund University 221 00 Lund Sweden
| | - Christelle N Prinz
- Division of Solid State Physics and NanoLund, Lund University 221 00 Lund Sweden
| |
Collapse
|
6
|
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
|
7
|
Baghery Saghchy Khorasani A, Yousefi AM, Bashash D. CAR NK cell therapy in hematologic malignancies and solid tumors; obstacles and strategies to overcome the challenges. Int Immunopharmacol 2022; 110:109041. [PMID: 35839565 DOI: 10.1016/j.intimp.2022.109041] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/29/2022] [Accepted: 07/07/2022] [Indexed: 02/08/2023]
Abstract
Adoptive cell treatment (ACT) utilizing chimeric antigen receptors (CAR) diverts the specificity of safe cells against a target-specific antigen and portrays exceptional potential for cancer treatment. While CAR T cell treatment has risen as a breakthrough with unprecedented results within the therapeutic procedures of human malignancies, different deficiencies including challenging and costly generation processes, strict patient qualification criteria, and undesirable toxicity have ruined its application. Unlike T cells, the application of natural killer (NK) cells has attracted consideration as a reasonable alternative owing to the major histocompatibility complex (MHC)-independency, shorter life expectancy, the potential to create an off-the-shelf immune product, and potent antitumor properties. In this article, we provide an updated review of the differences between CAR T and CAR NK cells, current enhancements in CAR NK design, the available sources for collecting NK cells, and strategies for the transduction step of the CARs to NK cells. Furthermore, we focus on the published and ongoing preclinical and clinical studies of CAR NK treatment strategies both in hematologic malignancies and solid tumors. We also discuss limitations and plausible solutions to improve the perseverance, function, safety, and efficacy of CAR NK cells with a special focus on solid tumors.
Collapse
Affiliation(s)
| | - Amir-Mohammad Yousefi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Muralidharan A, Pesch GR, Hubbe H, Rems L, Nouri-Goushki M, Boukany PE. Microtrap array on a chip for localized electroporation and electro-gene transfection. Bioelectrochemistry 2022; 147:108197. [PMID: 35810498 DOI: 10.1016/j.bioelechem.2022.108197] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/09/2022] [Accepted: 06/25/2022] [Indexed: 11/19/2022]
Abstract
We developed a localized single-cell electroporation chip to deliver exogenous biomolecules with high efficiency while maintaining high cell viability. In our microfluidic device, the cells are trapped in a microtrap array by flow, after which target molecules are supplied to the device and electrotransferred to the cells under electric pulses. The system provides the ability to monitor the electrotransfer of exogenous biomolecules in real time. We reveal through numerical simulations that localized electroporation is the mechanism of permeabilization in the microtrap array electroporation device. We demonstrate the simplicity and accuracy of this microtrap technology for electroporation by delivery of both small molecules using propidium iodide and large molecules using plasmid DNA for gene expression, illustrating the potential of this minimally invasive method to be widely used for precise intracellular delivery purposes (from bioprocess engineering to therapeutic applications).
Collapse
Affiliation(s)
- Aswin Muralidharan
- Department of Chemical Engineering, Delft University of Technology, van der Maasweg 9, 2629 HZ Delft, the Netherlands.
| | - Georg R Pesch
- Department of Chemical Engineering, Delft University of Technology, van der Maasweg 9, 2629 HZ Delft, the Netherlands
| | - Hendrik Hubbe
- Department of Chemical Engineering, Delft University of Technology, van der Maasweg 9, 2629 HZ Delft, the Netherlands
| | - Lea Rems
- Faculty of Electrical Engineering, University of Ljubljana, Trzaska 25, 1000 Ljubljana, Slovenia
| | - Mahdiyeh Nouri-Goushki
- Department of Chemical Engineering, Delft University of Technology, van der Maasweg 9, 2629 HZ Delft, the Netherlands
| | - Pouyan E Boukany
- Department of Chemical Engineering, Delft University of Technology, van der Maasweg 9, 2629 HZ Delft, the Netherlands.
| |
Collapse
|
9
|
Brooks JR, Mungloo I, Mirfendereski S, Quint JP, Paul D, Jaberi A, Park JS, Yang R. An equivalent circuit model for localized electroporation on porous substrates. Biosens Bioelectron 2022; 199:113862. [PMID: 34923307 PMCID: PMC8741749 DOI: 10.1016/j.bios.2021.113862] [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: 08/10/2021] [Revised: 11/29/2021] [Accepted: 12/02/2021] [Indexed: 11/02/2022]
Abstract
In vitro intracellular delivery is a fundamental challenge with no widely adopted methods capable of both delivering to millions of cells and controlling that delivery to a high degree of accuracy. One promising method is porous substrate electroporation (PSEP), where cells are cultured on porous substrates and electric fields are used to permeabilize discrete portions of the cell membrane for delivery. A major obstacle to the widespread use of PSEP is a poor understanding of the various impedances that constitute the system, including the impedances of the porous substrate and the cell monolayer, and how these impedances are influenced by experimental parameters. In response, we used impedance measurements to develop an equivalent circuit model that closely mimics the behavior of each of the main components of the PSEP system. This circuit model reveals for the first time the distribution of voltage across the electrode-electrolyte interface impedances, the channels of the porous substrate, the cell monolayer, and the transmembrane potential during PSEP. We applied sample waveforms through our model to understand how waveforms can be improved for future studies. Our model was validated from intracellular delivery of protein using PSEP.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ruiguo Yang
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA.
| |
Collapse
|
10
|
Duarte-Sanmiguel S, Panic A, Dodd DJ, Salazar-Puerta A, Moore JT, Lawrence WR, Nairon K, Francis C, Zachariah N, McCoy W, Turaga R, Skardal A, Carson WE, Higuita-Castro N, Gallego-Perez D. In Situ Deployment of Engineered Extracellular Vesicles into the Tumor Niche via Myeloid-Derived Suppressor Cells. Adv Healthc Mater 2022; 11:e2101619. [PMID: 34662497 PMCID: PMC8891033 DOI: 10.1002/adhm.202101619] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/26/2021] [Indexed: 12/19/2022]
Abstract
Extracellular vesicles (EVs) have emerged as a promising carrier system for the delivery of therapeutic payloads in multiple disease models, including cancer. However, effective targeting of EVs to cancerous tissue remains a challenge. Here, it is shown that nonviral transfection of myeloid-derived suppressor cells (MDSCs) can be leveraged to drive targeted release of engineered EVs that can modulate transfer and overexpression of therapeutic anticancer genes in tumor cells and tissue. MDSCs are immature immune cells that exhibit enhanced tropism toward tumor tissue and play a role in modulating tumor progression. Current MDSC research has been mostly focused on mitigating immunosuppression in the tumor niche; however, the tumor homing abilities of these cells present untapped potential to deliver EV therapeutics directly to cancerous tissue. In vivo and ex vivo studies with murine models of breast cancer show that nonviral transfection of MDSCs does not hinder their ability to home to cancerous tissue. Moreover, transfected MDSCs can release engineered EVs and mediate antitumoral responses via paracrine signaling, including decreased invasion/metastatic activity and increased apoptosis/necrosis. Altogether, these findings indicate that MDSCs can be a powerful tool for the deployment of EV-based therapeutics to tumor tissue.
Collapse
Affiliation(s)
| | - Ana Panic
- The Ohio State University, Department of Biomedical Engineering, Columbus, OH 43210
| | - Daniel J. Dodd
- The Ohio State University, Department of Biomedical Engineering, Columbus, OH 43210
- The Ohio State University, Biomedical Sciences Graduate Program, Columbus, OH 43210
| | - Ana Salazar-Puerta
- The Ohio State University, Department of Biomedical Engineering, Columbus, OH 43210
| | - Jordan T. Moore
- The Ohio State University, Department of Biomedical Engineering, Columbus, OH 43210
| | - William R. Lawrence
- The Ohio State University, Biomedical Sciences Graduate Program, Columbus, OH 43210
| | - Kylie Nairon
- The Ohio State University, Department of Biomedical Engineering, Columbus, OH 43210
| | - Carlie Francis
- The Ohio State University, Department of Biomedical Engineering, Columbus, OH 43210
| | - Natalie Zachariah
- The Ohio State University, Department of Biomedical Engineering, Columbus, OH 43210
| | - William McCoy
- The Ohio State University, Department of Biomedical Engineering, Columbus, OH 43210
| | - Rithvik Turaga
- The Ohio State University, Department of Biomedical Engineering, Columbus, OH 43210
| | - Aleksander Skardal
- The Ohio State University, Department of Biomedical Engineering, Columbus, OH 43210
| | - William E. Carson
- The Ohio State University, Department of Surgery, Columbus, OH 43210
| | - Natalia Higuita-Castro
- The Ohio State University, Department of Biomedical Engineering, Columbus, OH 43210
- The Ohio State University, Department of Surgery, Columbus, OH 43210
- The Ohio State University, Biophysics Program, OH 43210
| | - Daniel Gallego-Perez
- The Ohio State University, Department of Biomedical Engineering, Columbus, OH 43210
- The Ohio State University, Department of Surgery, Columbus, OH 43210
| |
Collapse
|
11
|
Schmidt D, Ebrahimabadi S, Gomes KRDS, de Moura Aguiar G, Cariati Tirapelle M, Nacasaki Silvestre R, de Azevedo JTC, Tadeu Covas D, Picanço-Castro V. Engineering CAR-NK cells: how to tune innate killer cells for cancer immunotherapy. IMMUNOTHERAPY ADVANCES 2022; 2:ltac003. [PMID: 35919494 PMCID: PMC9327111 DOI: 10.1093/immadv/ltac003] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 02/01/2022] [Indexed: 11/13/2022] Open
Abstract
Cell therapy is an innovative approach that permits numerous possibilities in the field of cancer treatment. CAR-T cells have been successfully used in patients with hematologic relapsed/refractory. However, the need for autologous sources for T cells is still a major drawback. CAR-NK cells have emerged as a promising resource using allogeneic cells that could be established as an off-the-shelf treatment. NK cells can be obtained from various sources, such as peripheral blood (PB), bone marrow, umbilical cord blood (CB), and induced pluripotent stem cells (iPSC), as well as cell lines. Genetic engineering of NK cells to express different CAR constructs for hematological cancers and solid tumors has shown promising preclinical results and they are currently being explored in multiple clinical trials. Several strategies have been employed to improve CAR-NK-cell expansion and cytotoxicity efficiency. In this article, we review the latest achievements and progress made in the field of CAR-NK-cell therapy.
Collapse
Affiliation(s)
- Dayane Schmidt
- Regional Blood Center of the School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
- Department of Medical Imaging, Hematology, and Clinical Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Sima Ebrahimabadi
- Regional Blood Center of the School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
- Department of Medical Imaging, Hematology, and Clinical Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Kauan Ribeiro de Sena Gomes
- Regional Blood Center of the School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
- Department of Medical Imaging, Hematology, and Clinical Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Graziela de Moura Aguiar
- Regional Blood Center of the School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Mariane Cariati Tirapelle
- Regional Blood Center of the School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
- Department of Medical Imaging, Hematology, and Clinical Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Renata Nacasaki Silvestre
- Regional Blood Center of the School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
- Department of Medical Imaging, Hematology, and Clinical Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Júlia Teixeira Cottas de Azevedo
- Regional Blood Center of the School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
- Department of Medical Imaging, Hematology, and Clinical Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Dimas Tadeu Covas
- Regional Blood Center of the School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
- Department of Medical Imaging, Hematology, and Clinical Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Virginia Picanço-Castro
- Regional Blood Center of the School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
12
|
Dong Z, Wang Y, Yin D, Hang X, Pu L, Zhang J, Geng J, Chang L. Advanced techniques for gene heterogeneity research: Single‐cell sequencing and on‐chip gene analysis systems. VIEW 2022. [DOI: 10.1002/viw.20210011] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Affiliation(s)
- Zaizai Dong
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering Beihang University Beijing China
| | - Yu Wang
- Department of Laboratory Medicine State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University/Collaborative Innovation Center Chengdu China
| | - Dedong Yin
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering Beihang University Beijing China
| | - Xinxin Hang
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering Beihang University Beijing China
| | - Lei Pu
- Department of Laboratory Medicine State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University/Collaborative Innovation Center Chengdu China
| | - Jianfu Zhang
- Department of Laboratory Medicine State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University/Collaborative Innovation Center Chengdu China
| | - Jia Geng
- Department of Laboratory Medicine State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University/Collaborative Innovation Center Chengdu China
| | - Lingqian Chang
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering Beihang University Beijing China
| |
Collapse
|
13
|
Dong Z, Chang L. Recent electroporation-based systems for intracellular molecule delivery. NANOTECHNOLOGY AND PRECISION ENGINEERING 2021. [DOI: 10.1063/10.0005649] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Zaizai Dong
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Lingqian Chang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| |
Collapse
|
14
|
Hosseini M, Habibi Z, Hosseini N, Abdoli S, Rezaei N. Preclinical studies of chimeric antigen receptor-modified natural killer cells in cancer immunotherapy: a review. Expert Opin Biol Ther 2021; 22:349-366. [PMID: 34541989 DOI: 10.1080/14712598.2021.1983539] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION As one of the most efficacious methods of cancer immunotherapy, chimeric antigen receptor-modified immune cells have recently drawn enormous attention. After the great success achieved with CAR-T-cells in cancer treatment both in preclinical setting and in the clinic, other types of immune cells, including natural killer (NK)-cells and macrophages, have been evaluated for their anti-cancer effects along with their potential superiority against CAR-T-cells, especially in terms of safety. First introduced by Tran et al. almost 26 years ago, CAR-NK-cells are now being considered as efficient immunotherapeutic modalities in various types of cancers, not only in preclinical setting but also in numerous phase I and II clinical studies. AREAS COVERED In this review, we aim to provide a comprehensive survey of the preclinical studies on CAR-NK-cells' development, with an evolutional approach on CAR structures and their associated signaling moieties. Current NK-cell sources and modes of gene transfer are also reviewed. EXPERT OPINION CAR-NK-cells have appeared as safe and effective immunotherapeutic tools in preclinical settings; however, designing CAR structures with an eye on their specific biology, along with choosing the optimal cell source and gene transfer method require further investigation to support clinical studies.
Collapse
Affiliation(s)
- Mina Hosseini
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Department of Pharmaceutical Biotechnology, School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Habibi
- School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Narges Hosseini
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Sina Abdoli
- School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Research Center for Immunodeficiencies (RCID), Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Abou-El-Enein M, Elsallab M, Feldman SA, Fesnak AD, Heslop HE, Marks P, Till BG, Bauer G, Savoldo B. Scalable Manufacturing of CAR T cells for Cancer Immunotherapy. Blood Cancer Discov 2021; 2:408-422. [PMID: 34568831 PMCID: PMC8462122 DOI: 10.1158/2643-3230.bcd-21-0084] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
As of April 2021, there are five commercially available chimeric antigen receptor (CAR) T cell therapies for hematological malignancies. With the current transition of CAR T cell manufacturing from academia to industry, there is a shift toward Good Manufacturing Practice (GMP)-compliant closed and automated systems to ensure reproducibility and to meet the increased demand for cancer patients. In this review we describe current CAR T cells clinical manufacturing models and discuss emerging technological advances that embrace scaling and production optimization. We summarize measures being used to shorten CAR T-cell manufacturing times and highlight regulatory challenges to scaling production for clinical use. Statement of Significance ∣ As the demand for CAR T cell cancer therapy increases, several closed and automated production platforms are being deployed, and others are in development.This review provides a critical appraisal of these technologies that can be leveraged to scale and optimize the production of next generation CAR T cells.
Collapse
Affiliation(s)
- Mohamed Abou-El-Enein
- Division of Medical Oncology, Department of Medicine, and Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Joint USC/CHLA Cell Therapy Program, University of Southern California, and Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Magdi Elsallab
- Joint USC/CHLA Cell Therapy Program, University of Southern California, and Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Steven A Feldman
- Stanford Center for Cancer Cell Therapy, Stanford Cancer Institute, Palo Alto, CA
| | - Andrew D Fesnak
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Helen E Heslop
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - Peter Marks
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Brian G Till
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Gerhard Bauer
- Institute for Regenerative Cures (IRC), University of California Davis, Sacramento, California, USA
| | - Barbara Savoldo
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
16
|
Chimeric Antigen Receptor-Engineered Natural Killer (CAR NK) Cells in Cancer Treatment; Recent Advances and Future Prospects. Stem Cell Rev Rep 2021; 17:2081-2106. [PMID: 34472037 PMCID: PMC8410173 DOI: 10.1007/s12015-021-10246-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/13/2021] [Indexed: 12/28/2022]
Abstract
Natural Killer (NK) cells are critical members of the innate immunity lymphocytes and have a critical role in host defense against malignant cells. Adoptive cell therapy (ACT) using chimeric antigen receptor (CAR) redirects the specificity of the immune cell against a target-specific antigen. ACT has recently created an outstanding opportunity for cancer treatment. Unlike CAR-armored T cells which hadnsome shortcomings as the CAR-receiving construct, Major histocompatibility complex (MHC)-independency, shorter lifespan, the potential to produce an off-the-shelf immune product, and potent anti-tumor properties of the NK cells has introduced NK cells as a potent alternative target for expression of CAR. Here, we aim to provide an updated overview on the current improvements in CAR NK design and immunobiology and describe the potential of CAR-modified NK cells as an alternative “off-the-shelf” carrier of CAR. We also provide lists for the sources of NK cells in the process of CAR NK cell production, different methods for transduction of the CAR genetic sequence to NK cells, the differences between CAR T and CAR NK, and CAR NK-targeted tumor antigens in current studies. Additionally, we provide data on recently published preclinical and clinical studies of CAR NK therapy and a list of finished and ongoing clinical trials. For achieving CAR NK products with higher efficacy and safety, we discuss current challenges in transduction and expansion of CAR NK cells, CAR NK therapy side effects, and challenges that limit the optimal efficacy of CAR NK cells and recommend possible solutions to enhance the persistence, function, safety, and efficacy of CAR NK cells with a special focus on solid tumors.
Collapse
|
17
|
Tay A, Melosh N. Mechanical Stimulation after Centrifuge-Free Nano-Electroporative Transfection Is Efficient and Maintains Long-Term T Cell Functionalities. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2103198. [PMID: 34396686 PMCID: PMC8475193 DOI: 10.1002/smll.202103198] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/05/2021] [Indexed: 05/08/2023]
Abstract
Transfection is an essential step in genetic engineering and cell therapies. While a number of non-viral micro- and nano-technologies have been developed to deliver DNA plasmids into the cell cytoplasm, one of the most challenging and least efficient steps is DNA transport to and expression in the nucleus. Here, the magnetic nano-electro-injection (MagNEI) platform is described which makes use of oscillatory mechanical stimulation after cytoplasmic delivery with high aspect-ratio nano-structures to achieve stable (>2 weeks) net transfection efficiency (efficiency × viability) of 50% in primary human T cells. This is, to the best of the authors' knowledge, the highest net efficiency reported for primary T cells using a centrifuge-free, non-viral transfection method, in the absence of cell selection, and with a clinically relevant cargo size (>12 kbp). Wireless mechanical stimulation downregulates the expression of microtubule motor protein gene, KIF2A, which increases local DNA concentration near the nuclei, resulting in enhanced DNA transfection. Magnetic forces also accelerate membrane repair by promoting actin cytoskeletal remodeling which preserves key biological attributes including cell proliferation and gene expressions. These results demonstrate MagNEI as a powerful non-viral transfection technique for progress toward fully closed, end-to-end T cell manufacturing with less human labor, lower production cost, and shorter delay.
Collapse
Affiliation(s)
- Andy Tay
- Department of Biomedical Engineering, National University of Singapore, Singapore 117583
- Institute of Health Innovation & Technology, National University of Singapore, Singapore 117599
| | - Nicholas Melosh
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305
| |
Collapse
|
18
|
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: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [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
|
19
|
Dong Z, Yan S, Liu B, Hao Y, Lin L, Chang T, Sun H, Wang Y, Li H, Wu H, Hang X, He S, Hu J, Xue X, Wu N, Chang L. Single Living Cell Analysis Nanoplatform for High-Throughput Interrogation of Gene Mutation and Cellular Behavior. NANO LETTERS 2021; 21:4878-4886. [PMID: 33830766 DOI: 10.1021/acs.nanolett.1c00199] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The genetic heterogeneities in cancer cells pose challenges to achieving precise drug treatment in a widely applicable manner. Most single-cell gene analysis methods rely on cell lysis for gene extraction and identification, showing limited capacity to provide the correlation of genetic properties and real-time cellular behaviors. Here, we report a single living cell analysis nanoplatform that enables interrogating gene properties and drug resistance in millions of single cells. We designed a Domino-probe to identify intracellular target RNAs while releasing 10-fold amplified fluorescence signals. An on-chip addressable microwell-nanopore array was developed for enhanced electro-delivery of the Domino-probe and in situ observation of cell behaviors. The proof-of-concept of the system was validated in primary lung cancer cell samples, revealing the positive-correlation of the ratio of EGFR mutant cells with their drug susceptibilities. This platform provides a high-throughput yet precise tool for exploring the relationship between intracellular genes and cell behaviors at the single-cell level.
Collapse
Affiliation(s)
- Zaizai Dong
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Shi Yan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Bing Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yongcun Hao
- Ministry of Education Key Laboratory of Micro and Nano Systems for Aerospace, Northwestern Polytechnical University, Xi'an 710072, China
| | - Long Lin
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Tianrui Chang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Hong Sun
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Yusen Wang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Hu Li
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Han Wu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Xinxin Hang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Shiqi He
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Jiaming Hu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Xinying Xue
- Department of Respiratory and Critical Care, Beijing Shijitan Hospital, Capital Medical University, Peking University Ninth School of Clinical Medicine, Beijing 100038, China
- Department of Respiratory and Critical Care, Chinese PLA General Hospital, Beijing 100853, China
- Affiliated Hospital of Weifang Medical University, Shandong 261031, China
| | - Nan Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Lingqian Chang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| |
Collapse
|
20
|
Kumar ARK, Shou Y, Chan B, L K, Tay A. Materials for Improving Immune Cell Transfection. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2007421. [PMID: 33860598 DOI: 10.1002/adma.202007421] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/08/2020] [Indexed: 06/12/2023]
Abstract
Chimeric antigen receptor T cell (CAR-T) therapy holds great promise for preventing and treating deadly diseases such as cancer. However, it remains challenging to transfect and engineer primary immune cells for clinical cell manufacturing. Conventional tools using viral vectors and bulk electroporation suffer from low efficiency while posing risks like viral transgene integration and excessive biological perturbations. Emerging techniques using microfluidics, nanoparticles, and high-aspect-ratio nanostructures can overcome these challenges, and on top of that, provide universal and high-throughput cargo delivery. Herein, the strengths and limitations of traditional and emerging materials for immune cell transfection, and commercial development of these tools, are discussed. To enhance the characterization of transfection techniques and uptake by the clinical community, a list of in vitro and in vivo assays to perform, along with relevant protocols, is recommended. The overall aim, herein, is to motivate the development of novel materials to meet rising demand in transfection for clinical CAR-T cell manufacturing.
Collapse
Affiliation(s)
- Arun R K Kumar
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
| | - Yufeng Shou
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Brian Chan
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Krishaa L
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Andy Tay
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
| |
Collapse
|
21
|
Sarno B, Heineck D, Heller MJ, Ibsen SD. Dielectrophoresis: Developments and applications from 2010 to 2020. Electrophoresis 2021; 42:539-564. [PMID: 33191521 PMCID: PMC7986072 DOI: 10.1002/elps.202000156] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 09/22/2020] [Accepted: 10/21/2020] [Indexed: 12/19/2022]
Abstract
The 20th century has seen tremendous innovation of dielectrophoresis (DEP) technologies, with applications being developed in areas ranging from industrial processing to micro- and nanoscale biotechnology. From 2010 to present day, there have been 981 publications about DEP. Of over 2600 DEP patents held by the United States Patent and Trademark Office, 106 were filed in 2019 alone. This review focuses on DEP-based technologies and application developments between 2010 and 2020, with an aim to highlight the progress and to identify potential areas for future research. A major trend over the last 10 years has been the use of DEP techniques for biological and clinical applications. It has been used in various forms on a diverse array of biologically derived molecules and particles to manipulate and study them including proteins, exosomes, bacteria, yeast, stem cells, cancer cells, and blood cells. DEP has also been used to manipulate nano- and micron-sized particles in order to fabricate different structures. The next 10 years are likely to see the increase in DEP-related patent applications begin to result in a greater level of technology commercialization. Also during this time, innovations in DEP technology will likely be leveraged to continue the existing trend to further biological and medical-focused applications as well as applications in microfabrication. As a tool leveraged by engineering and imaginative scientific design, DEP offers unique capabilities to manipulate small particles in precise ways that can help solve problems and enable scientific inquiry that cannot be addressed using conventional methods.
Collapse
Affiliation(s)
- Benjamin Sarno
- Oregon Health and Science University–The Knight Cancer Institute's Cancer Early Detection Advanced Research CenterPortlandORUSA
- University of California San Diego–NanoengineeringLa JollaCAUSA
| | - Daniel Heineck
- Oregon Health and Science University–The Knight Cancer Institute's Cancer Early Detection Advanced Research CenterPortlandORUSA
| | - Michael J. Heller
- Oregon Health and Science University–The Knight Cancer Institute's Cancer Early Detection Advanced Research CenterPortlandORUSA
- University of California San Diego–NanoengineeringLa JollaCAUSA
| | - Stuart D. Ibsen
- Oregon Health and Science University–The Knight Cancer Institute's Cancer Early Detection Advanced Research CenterPortlandORUSA
- Oregon Health and Science University–Biomedical EngineeringPortlandORUSA
| |
Collapse
|
22
|
Ghaemi A, Bagheri E, Abnous K, Taghdisi SM, Ramezani M, Alibolandi M. CRISPR-cas9 genome editing delivery systems for targeted cancer therapy. Life Sci 2020; 267:118969. [PMID: 33385410 DOI: 10.1016/j.lfs.2020.118969] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 02/07/2023]
Abstract
The prokaryotic CRISPR-Cas systems could be applied as revolutionized genome editing tool in live cells of various species to modify, visualize and identify definite sequences of DNA and RNA. CRISPR-Cas could edit the genome by homology-directed repair and non-homologous end joining mechanisms. Furthermore, DNA-targeting modification by CRISPR-Cas methodology provides opportunity for diagnosis, therapy and the genetic disorders investigation. Here, we summarized delivery systems employed for CRISPR-Cas9 for genome editing. Then preclinical studies of the CRISPR-Cas9-based therapeutics will be discussed considering the associated challenges and developments in its translation to clinic for cancer therapy.
Collapse
Affiliation(s)
- Asma Ghaemi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elnaz Bagheri
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mohammad Taghdisi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
23
|
Yilmaz A, Cui H, Caligiuri MA, Yu J. Chimeric antigen receptor-engineered natural killer cells for cancer immunotherapy. J Hematol Oncol 2020; 13:168. [PMID: 33287875 PMCID: PMC7720606 DOI: 10.1186/s13045-020-00998-9] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 11/12/2020] [Indexed: 12/13/2022] Open
Abstract
Natural killer (NK) cells are a critical component of the innate immune system. Chimeric antigen receptors (CARs) re-direct NK cells toward tumor cells carrying corresponding antigens, creating major opportunities in the fight against cancer. CAR NK cells have the potential for use as universal CAR cells without the need for human leukocyte antigen matching or prior exposure to tumor-associated antigens. Exciting data from recent clinical trials have renewed interest in the field of cancer immunotherapy due to the potential of CAR NK cells in the production of "off-the-shelf" anti-cancer immunotherapeutic products. Here, we provide an up-to-date comprehensive overview of the recent advancements in key areas of CAR NK cell research and identify under-investigated research areas. We summarize improvements in CAR design and structure, advantages and disadvantages of using CAR NK cells as an alternative to CAR T cell therapy, and list sources to obtain NK cells. In addition, we provide a list of tumor-associated antigens targeted by CAR NK cells and detail challenges in expanding and transducing NK cells for CAR production. We additionally discuss barriers to effective treatment and suggest solutions to improve CAR NK cell function, proliferation, persistence, therapeutic effectiveness, and safety in solid and liquid tumors.
Collapse
Affiliation(s)
- Ahmet Yilmaz
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Hanwei Cui
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Michael A Caligiuri
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, 1500 E. Duarte Road, KCRB, Bldg. 158, 3rd Floor, Room 3017, Los Angeles, CA, 91010, USA
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Los Angeles, CA, 91010, USA
- Department of Immuno-Oncology, City of Hope Beckman Research Institute, Los Angeles, CA, 91010, USA
- City of Hope Comprehensive Cancer Center and Beckman Research Institute, Los Angeles, CA, 91010, USA
| | - Jianhua Yu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, 1500 E. Duarte Road, KCRB, Bldg. 158, 3rd Floor, Room 3017, Los Angeles, CA, 91010, USA.
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Los Angeles, CA, 91010, USA.
- Department of Immuno-Oncology, City of Hope Beckman Research Institute, Los Angeles, CA, 91010, USA.
- City of Hope Comprehensive Cancer Center and Beckman Research Institute, Los Angeles, CA, 91010, USA.
| |
Collapse
|
24
|
Brooks J, Minnick G, Mukherjee P, Jaberi A, Chang L, Espinosa HD, Yang R. High Throughput and Highly Controllable Methods for In Vitro Intracellular Delivery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2004917. [PMID: 33241661 PMCID: PMC8729875 DOI: 10.1002/smll.202004917] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/06/2020] [Indexed: 05/03/2023]
Abstract
In vitro and ex vivo intracellular delivery methods hold the key for releasing the full potential of tissue engineering, drug development, and many other applications. In recent years, there has been significant progress in the design and implementation of intracellular delivery systems capable of delivery at the same scale as viral transfection and bulk electroporation but offering fewer adverse outcomes. This review strives to examine a variety of methods for in vitro and ex vivo intracellular delivery such as flow-through microfluidics, engineered substrates, and automated probe-based systems from the perspective of throughput and control. Special attention is paid to a particularly promising method of electroporation using micro/nanochannel based porous substrates, which expose small patches of cell membrane to permeabilizing electric field. Porous substrate electroporation parameters discussed include system design, cells and cargos used, transfection efficiency and cell viability, and the electric field and its effects on molecular transport. The review concludes with discussion of potential new innovations which can arise from specific aspects of porous substrate-based electroporation platforms and high throughput, high control methods in general.
Collapse
Affiliation(s)
- Justin Brooks
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Grayson Minnick
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Prithvijit Mukherjee
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Arian Jaberi
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Lingqian Chang
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Horacio D. Espinosa
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Theoretical and Applied Mechanics Program, Northwestern University, Evanston, IL, 60208, USA
| | - Ruiguo Yang
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| |
Collapse
|
25
|
Duckert B, Vinkx S, Braeken D, Fauvart M. Single-cell transfection technologies for cell therapies and gene editing. J Control Release 2020; 330:963-975. [PMID: 33160005 DOI: 10.1016/j.jconrel.2020.10.068] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/30/2020] [Accepted: 10/31/2020] [Indexed: 12/29/2022]
Abstract
Advances in gene editing and cell therapies have recently led to outstanding clinical successes. However, the lack of a cost-effective manufacturing process prevents the democratization of these innovative medical tools. Due to the common use of viral vectors, the step of transfection in which cells are engineered to gain new functions, is a major bottleneck in making safe and affordable cell products. A promising opportunity lies in Single-Cell Transfection Technologies (SCTTs). SCTTs have demonstrated higher efficiency, safety and scalability than conventional transfection methods. They can also feature unique abilities such as substantial dosage control over the cargo delivery, single-cell addressability and integration in microdevices comprising multiple monitoring modalities. Unfortunately, the potential of SCTTs is not fully appreciated: they are most often restricted to research settings with little adoption in clinical settings. To encourage their adoption, we review and compare recent developments in SCTTs, and how they can enable selected clinical applications. To help bridge the gap between fundamental research and its translation to the clinic, we also describe how Good Manufacturing Practices (GMP) can be integrated in the design of SCTTs.
Collapse
Affiliation(s)
- Bastien Duckert
- Department of Physics and Astronomy, KU Leuven, Celestijnenlaan 200d, 3001 Leuven, Belgium; IMEC, Kapeldreef 75, 3001 Leuven, Belgium.
| | | | | | | |
Collapse
|
26
|
Moore JT, Wier CG, Lemmerman LR, Ortega-Pineda L, Dodd DJ, Lawrence WR, Duarte-Sanmiguel S, Dathathreya K, Diaz-Starokozheva L, Harris HN, Sen CK, Valerio IL, Higuita-Castro N, Arnold WD, Kolb SJ, Gallego-Perez D. Nanochannel-Based Poration Drives Benign and Effective Nonviral Gene Delivery to Peripheral Nerve Tissue. ADVANCED BIOSYSTEMS 2020; 4:e2000157. [PMID: 32939985 PMCID: PMC7704786 DOI: 10.1002/adbi.202000157] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/04/2020] [Accepted: 08/18/2020] [Indexed: 01/01/2023]
Abstract
While gene and cell therapies have emerged as promising treatment strategies for various neurological conditions, heavy reliance on viral vectors can hamper widespread clinical implementation. Here, the use of tissue nanotransfection as a platform nanotechnology to drive nonviral gene delivery to nerve tissue via nanochannels, in an effective, controlled, and benign manner is explored. TNT facilitates plasmid DNA delivery to the sciatic nerve of mice in a voltage-dependent manner. Compared to standard bulk electroporation (BEP), impairment in toe-spread and pinprick response is not caused by TNT, and has limited to no impact on electrophysiological parameters. BEP, however, induces significant nerve damage and increases macrophage immunoreactivity. TNT is subsequently used to deliver vasculogenic cell therapies to crushed nerves via delivery of reprogramming factor genes Etv2, Foxc2, and Fli1 (EFF). The results indicate the TNT-based delivery of EFF in a sciatic nerve crush model leads to increased vascularity, reduced macrophage infiltration, and improved recovery in electrophysiological parameters compared to crushed nerves that are TNT-treated with sham/empty plasmids. Altogether, the results indicate that TNT can be a powerful platform nanotechnology for localized nonviral gene delivery to nerve tissue, in vivo, and the deployment of reprogramming-based cell therapies for nerve repair/regeneration.
Collapse
Affiliation(s)
- Jordan T. Moore
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
| | | | - Luke R. Lemmerman
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
| | | | - Daniel J. Dodd
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, USA
| | - William R. Lawrence
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Silvia Duarte-Sanmiguel
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
- Department of Human Sciences, The Ohio State University, Columbus, OH, USA
| | - Kavya Dathathreya
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
| | | | - Hallie N. Harris
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Chandan K. Sen
- School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Ian L. Valerio
- Plastic and Reconstructive Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Natalia Higuita-Castro
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - W. David Arnold
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Stephen J. Kolb
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Biological Chemistry and Pharmacology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Daniel Gallego-Perez
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
27
|
Chang L, Chitrakar C, Nouri M. 3D Nanochannel Electroporation for Macromolecular Nucleotide Delivery. Methods Mol Biol 2020; 2050:69-77. [PMID: 31468480 DOI: 10.1007/978-1-4939-9740-4_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Delivery of macromolecular nucleotides into the living cells holds a great promise for the development of new therapeutics. However, its abilities for adoptive immunotherapy, cell reprogramming, and primary cell transfection have been long-term hindered by the lack of a system that can locally deliver engineered therapeutic nucleotides (e.g., plasmids, siRNAs, miRNAs) without causing any side effects. In this chapter, the performance of a novel 3D nanoelectroporation system (3D NEP) is highlighted in three scenarios-adoptive immunotherapy, cell reprogramming, and adult mouse primary cardiomyocyte transfection. Detailed protocols were given to introduce the 3D NEP system assembly, as well as their applications in (1) natural killer (NK) cells transfection by delivery of chimeric antigen receptor (CAR) plasmids; (2) mouse embryonic fibroblasts transfection with OSKM factors; and (3) miR-29b molecular beacon (BMs) delivery into primary cardiomyocytes for interrogating the side effect of miR-29b-assisted treatment.
Collapse
Affiliation(s)
- Lingqian Chang
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China.
- Institute of Nanotechnology for Single Cell Analysis (INSCA), Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China.
| | - Chandani Chitrakar
- Department of Biomedical Engineering, University of North Texas, Denton, TX, USA
| | - Mehdi Nouri
- Department of Biomedical Engineering, University of North Texas, Denton, TX, USA
| |
Collapse
|
28
|
Liu L, Yang C, Liu C, Piao J, Kaw HY, Cui J, Shang H, Ri HC, Kim JM, Jin M, Li D. Open-tubular radially cyclical electric field-flow fractionation (OTR-CyElFFF): an online concentric distribution strategy for simultaneous separation of microparticles. LAB ON A CHIP 2020; 20:3535-3543. [PMID: 32852497 DOI: 10.1039/d0lc00620c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
An open-tubular radially cyclical electric field-flow fractionation technique which achieves the online separation of microparticles in a functional annular channel is proposed in this study. The system was set up by using a stainless steel tube and a platinum wire modified with ionic liquid/mesoporous silica materials as the external and internal electrodes. The feasibility for online separation of various particles was experimentally demonstrated. Particles in the channel were affected by a radial electric field and field-flow fractionation (FFF). On the cross section, different particles showed distinctive migration distances depending on their own properties and the different magnitudes of forces being exerted. The same kind of particles form an annular distribution within the same annulus while different particles form annular distributions at varied concentric annuli through electrophoresis. Under a laminar flow of FFF, different sizes of particles formed a conical arrangement within the annular separation channel. With the joint influence of electric field and flow field, different trajectories were obtained and the particles were eventually separated. Voltage, frequency and duty cycle value are the main parameters affecting the separation of particles. By adjusting these parameters, particles migrate in a zigzag trajectory on one side of the electrodes (mode I) and reach both sides of the electrodes (mode II). Six polystyrene particles were completely separated with high resolution within several minutes. Our system offers numerous advantages of label-free, high-resolution and online separation without tedious operations, and it is a promising tool for the effective separation of various micro-objects.
Collapse
Affiliation(s)
- Lu Liu
- Department of Chemistry, Yanbian University, Park Road 977, Yanji City, Jilin Province 133002, PR China.
| | - Cui Yang
- Department of Chemistry, Changchun Normal University, Changji North Road 677, Changchun City, Jilin Province 130032, China
| | - Cuicui Liu
- Department of Chemistry, Yanbian University, Park Road 977, Yanji City, Jilin Province 133002, PR China.
| | - Jishou Piao
- Department of Chemistry, Yanbian University, Park Road 977, Yanji City, Jilin Province 133002, PR China.
| | - Han Yeong Kaw
- Department of Chemistry, Yanbian University, Park Road 977, Yanji City, Jilin Province 133002, PR China.
| | - Jiaxuan Cui
- Department of Chemistry, Yanbian University, Park Road 977, Yanji City, Jilin Province 133002, PR China.
| | - Haibo Shang
- Department of Chemistry, Yanbian University, Park Road 977, Yanji City, Jilin Province 133002, PR China.
| | - Hyok Chol Ri
- Department of Chemistry, Yanbian University, Park Road 977, Yanji City, Jilin Province 133002, PR China.
| | - Ji Man Kim
- Department of Chemistry, Sungkyunkwan University, Korea
| | - Mingshi Jin
- Department of Chemistry, Yanbian University, Park Road 977, Yanji City, Jilin Province 133002, PR China.
| | - Donghao Li
- Department of Chemistry, Yanbian University, Park Road 977, Yanji City, Jilin Province 133002, PR China.
| |
Collapse
|
29
|
Benhal P, Quashie D, Kim Y, Ali J. Insulator Based Dielectrophoresis: Micro, Nano, and Molecular Scale Biological Applications. SENSORS (BASEL, SWITZERLAND) 2020; 20:E5095. [PMID: 32906803 PMCID: PMC7570478 DOI: 10.3390/s20185095] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 08/16/2020] [Accepted: 09/04/2020] [Indexed: 12/31/2022]
Abstract
Insulator based dielectrophoresis (iDEP) is becoming increasingly important in emerging biomolecular applications, including particle purification, fractionation, and separation. Compared to conventional electrode-based dielectrophoresis (eDEP) techniques, iDEP has been demonstrated to have a higher degree of selectivity of biological samples while also being less biologically intrusive. Over the past two decades, substantial technological advances have been made, enabling iDEP to be applied from micro, to nano and molecular scales. Soft particles, including cell organelles, viruses, proteins, and nucleic acids, have been manipulated using iDEP, enabling the exploration of subnanometer biological interactions. Recent investigations using this technique have demonstrated a wide range of applications, including biomarker screening, protein folding analysis, and molecular sensing. Here, we review current state-of-art research on iDEP systems and highlight potential future work.
Collapse
Affiliation(s)
- Prateek Benhal
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, FL 32310, USA;
- National High Magnetic Field Laboratory, Tallahassee, FL 32310, USA
| | - David Quashie
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, FL 32310, USA;
- National High Magnetic Field Laboratory, Tallahassee, FL 32310, USA
| | - Yoontae Kim
- American Dental Association Science & Research Institute, Gaithersburg, MD 20899, USA;
| | - Jamel Ali
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, FL 32310, USA;
- National High Magnetic Field Laboratory, Tallahassee, FL 32310, USA
| |
Collapse
|
30
|
Tay A. The Benefits of Going Small: Nanostructures for Mammalian Cell Transfection. ACS NANO 2020; 14:7714-7721. [PMID: 32631053 DOI: 10.1021/acsnano.0c04624] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Nanostructures, with their localized interactions with mammalian cells, can offer better efficiency and lower cell perturbation than conventional viral, biochemical, and electroporation transfection techniques. In this Perspective, I describe the different stages of transfection and provide a comparison of transfection techniques based on their mechanisms. Focusing on specific aims of transfection, I also illustrate how recent developments in high-aspect-ratio nanostructures have endowed them with properties that are superior to existing viral, biochemical, and electroporation methods as a versatile technique to deliver a variety of cargoes and to interface with different mammalian cell types for biomedical applications. Finally, I describe the challenges associated with transfection that need to be overcome to enhance cargo delivery efficiency and clinical translation.
Collapse
Affiliation(s)
- Andy Tay
- Department of Biomedical Engineering, National University of Singapore, Singapore 117583
| |
Collapse
|
31
|
Zhang Z, Luo Y, Nie X, Yu D, Xing X. A one-step molded microfluidic chip featuring a two-layer silver-PDMS microelectrode for dielectrophoretic cell separation. Analyst 2020; 145:5603-5614. [PMID: 32776070 DOI: 10.1039/d0an01085e] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Dielectrophoresis (DEP) is a powerful technique for label-free cell separation in microfluidics. Easily-fabricated DEP separators with low cost and short turnaround time are in extremely high demand in practical applications, especially clinical usage where disposable devices are needed. DEP separators exploiting microelectrodes made of conducting polydimethylsiloxane (PDMS) composites enable the construction of advantageous 3D volumetric electrodes with a simple soft-lithography process. Yet, existing devices incorporating microelectrodes in conducting PDMS generally have their fluidic sidewalls constructed using a different material, and consequently require extra lithography of a sacrificial layer on the semi-finished master for molding the electrode and fluidic sidewalls in separate steps. Here we demonstrate a novel microfluidic DEP separator with a 3D electrode and fluidic structure entirely integrated within silver-PDMS composites. We develop a further simplified one-step molding process with lower cost using a readily-available and reusable SU8 master, eliminating the need for the additional lithography step in existing techniques. The uniquely designed two-layer electrode exhibits a spatially non-uniform electric field that enables cell migration in the vertical direction. The electrode upper layer then offers a harbor-like region for the trapping of the target cells that have drifted upwards, which shelters them from being dragged away by the main flow streams in the lower layer, and thus allows higher operation flow rate. We also optimize the upper layer thickness as a critical dimension for protecting the trapped cells from high drag and show easy widening of our device by elongation of the digits. We demonstrate that the elongated digits involving more parallel flow paths maintain a high capture efficiency of 95.4% for live cells with 85.6% purity in the separation of live/dead HeLa cells. We also investigate the device feasibility in a viability assay for cells post anti-cancer drug treatment.
Collapse
Affiliation(s)
- Zhongle Zhang
- College of Information Science and Technology, Beijing University of Chemical Technology, No. 15 North 3rd Ring Rd., Beijing, 100029, China.
| | | | | | | | | |
Collapse
|
32
|
Early Intervention in Ischemic Tissue with Oxygen Nanocarriers Enables Successful Implementation of Restorative Cell Therapies. Cell Mol Bioeng 2020; 13:435-446. [PMID: 33184576 DOI: 10.1007/s12195-020-00621-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 05/20/2020] [Indexed: 01/01/2023] Open
Abstract
Background Tissue ischemia contributes to necrosis and infection. While angiogenic cell therapies have emerged as a promising strategy against ischemia, current approaches to cell therapies face multiple hurdles. Recent advances in nuclear reprogramming could potentially overcome some of these limitations. However, under severely ischemic conditions necrosis could outpace reprogramming-based repair. As such, adjunctive measures are required to maintain a minimum level of tissue viability/activity for optimal response to restorative interventions. Methods Here we explored the combined use of polymerized hemoglobin (PolyHb)-based oxygen nanocarriers with Tissue Nano-Transfection (TNT)-driven restoration to develop tissue preservation/repair strategies that could potentially be used as a first line of care. Random-pattern cutaneous flaps were created in a mouse model of ischemic injury. PolyHbs with high and low oxygen affinity were synthesized and injected into the tissue flap at various timepoints of ischemic injury. The degree of tissue preservation was evaluated in terms of perfusion, oxygenation, and resulting necrosis. TNT was then used to deploy reprogramming-based vasculogenic cell therapies to the flaps via nanochannels. Reprogramming/repair outcomes were evaluated in terms of vascularity and necrosis. Results Flaps treated with PolyHbs exhibited a gradual decrease in necrosis as a function of time-to-intervention, with low oxygen affinity PolyHb showing the best outcomes. TNT-based intervention of the flap in combination with PolyHb successfully curtailed advanced necrosis compared to flaps treated with only PolyHb or TNT alone. Conclusions These results indicate that PolyHb and TNT technologies could potentially be synergistically deployed and used as early intervention measures to combat severe tissue ischemia.
Collapse
|
33
|
Phenotypic Plasticity of Invasive Edge Glioma Stem-like Cells in Response to Ionizing Radiation. Cell Rep 2020; 26:1893-1905.e7. [PMID: 30759398 PMCID: PMC6594377 DOI: 10.1016/j.celrep.2019.01.076] [Citation(s) in RCA: 151] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 10/12/2018] [Accepted: 01/18/2019] [Indexed: 12/23/2022] Open
Abstract
Unresectable glioblastoma (GBM) cells in the invading tumor edge can act as seeds for recurrence. The molecular and
phenotypic properties of these cells remain elusive. Here, we report that the invading edge and tumor core have two distinct types
of glioma stem-like cells (GSCs) that resemble proneural (PN) and mesenchymal (MES) subtypes, respectively. Upon exposure to
ionizing radiation (IR), GSCs, initially enriched for a CD133+ PN signature, transition to a CD109+ MES
subtype in a C/EBP-β-dependent manner. Our gene expression analysis of paired cohorts of patients with primary and
recurrent GBMs identified a CD133-to-CD109 shift in tumors with an MES recurrence. Patient-derived
CD133−/CD109+ cells are highly enriched with clonogenic, tumor-initiating, and
radiation-resistant properties, and silencing CD109 significantly inhibits these phenotypes. We also report a conserved regulation
of YAP/TAZ pathways by CD109 that could be a therapeutic target in GBM. Minata et al., in response to the proinflammatory environment induced by radiation, find that the tumor cells at the
invasive edge acquire the expression of the CD109 protein concomitantly losing CD133. CD109 drives oncogenic signaling through the
YAP/TAZ pathway, confers radioresistance to the cells, and represents a new potential therapeutic target for glioblastoma.
Collapse
|
34
|
Fang J, Hsueh YY, Soto J, Sun W, Wang J, Gu Z, Khademhosseini A, Li S. Engineering Biomaterials with Micro/Nanotechnologies for Cell Reprogramming. ACS NANO 2020; 14:1296-1318. [PMID: 32011856 PMCID: PMC10067273 DOI: 10.1021/acsnano.9b04837] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Cell reprogramming is a revolutionized biotechnology that offers a powerful tool to engineer cell fate and function for regenerative medicine, disease modeling, drug discovery, and beyond. Leveraging advances in biomaterials and micro/nanotechnologies can enhance the reprogramming performance in vitro and in vivo through the development of delivery strategies and the control of biophysical and biochemical cues. In this review, we present an overview of the state-of-the-art technologies for cell reprogramming and highlight the recent breakthroughs in engineering biomaterials with micro/nanotechnologies to improve reprogramming efficiency and quality. Finally, we discuss future directions and challenges for reprogramming technologies and clinical translation.
Collapse
Affiliation(s)
- Jun Fang
- Department of Bioengineering , University of California, Los Angeles , Los Angeles , California 90095 , United States
- Department of Medicine , University of California, Los Angeles , Los Angeles , California 90095 , United States
| | - Yuan-Yu Hsueh
- Department of Bioengineering , University of California, Los Angeles , Los Angeles , California 90095 , United States
- Division of Plastic Surgery, Department of Surgery, College of Medicine , National Cheng Kung University Hospital , Tainan 70456 , Taiwan
| | - Jennifer Soto
- Department of Bioengineering , University of California, Los Angeles , Los Angeles , California 90095 , United States
- Department of Medicine , University of California, Los Angeles , Los Angeles , California 90095 , United States
| | - Wujin Sun
- Department of Bioengineering , University of California, Los Angeles , Los Angeles , California 90095 , United States
- Center for Minimally Invasive Therapeutics (C-MIT), California NanoSystems Institute , University of California, Los Angeles , Los Angles , California 90095 , United States
| | - Jinqiang Wang
- Department of Bioengineering , University of California, Los Angeles , Los Angeles , California 90095 , United States
- Center for Minimally Invasive Therapeutics (C-MIT), California NanoSystems Institute , University of California, Los Angeles , Los Angles , California 90095 , United States
| | - Zhen Gu
- Department of Bioengineering , University of California, Los Angeles , Los Angeles , California 90095 , United States
- Center for Minimally Invasive Therapeutics (C-MIT), California NanoSystems Institute , University of California, Los Angeles , Los Angles , California 90095 , United States
- Jonsson Comprehensive Cancer Center , University of California, Los Angeles , Los Angeles , California 90095 , United States
| | - Ali Khademhosseini
- Department of Bioengineering , University of California, Los Angeles , Los Angeles , California 90095 , United States
- Center for Minimally Invasive Therapeutics (C-MIT), California NanoSystems Institute , University of California, Los Angeles , Los Angles , California 90095 , United States
- Department of Chemical and Biomolecular Engineering , University of California, Los Angeles , Los Angeles , California 90095 , United States
- Department of Radiology , University of California, Los Angeles , Los Angeles , California 90095 , United States
| | - Song Li
- Department of Bioengineering , University of California, Los Angeles , Los Angeles , California 90095 , United States
- Department of Medicine , University of California, Los Angeles , Los Angeles , California 90095 , United States
- Center for Minimally Invasive Therapeutics (C-MIT), California NanoSystems Institute , University of California, Los Angeles , Los Angles , California 90095 , United States
| |
Collapse
|
35
|
Dixit HG, Starr R, Dundon ML, Pairs PI, Yang X, Zhang Y, Nampe D, Ballas CB, Tsutsui H, Forman SJ, Brown CE, Rao MP. Massively-Parallelized, Deterministic Mechanoporation for Intracellular Delivery. NANO LETTERS 2020; 20:860-867. [PMID: 31647675 PMCID: PMC8210888 DOI: 10.1021/acs.nanolett.9b03175] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Microfluidic intracellular delivery approaches based on plasma membrane poration have shown promise for addressing the limitations of conventional cellular engineering techniques in a wide range of applications in biology and medicine. However, the inherent stochasticity of the poration process in many of these approaches often results in a trade-off between delivery efficiency and cellular viability, thus potentially limiting their utility. Herein, we present a novel microfluidic device concept that mitigates this trade-off by providing opportunity for deterministic mechanoporation (DMP) of cells en masse. This is achieved by the impingement of each cell upon a single needle-like penetrator during aspiration-based capture, followed by diffusive influx of exogenous cargo through the resulting membrane pore, once the cells are released by reversal of flow. Massive parallelization enables high throughput operation, while single-site poration allows for delivery of small and large-molecule cargos in difficult-to-transfect cells with efficiencies and viabilities that exceed both conventional and emerging transfection techniques. As such, DMP shows promise for advancing cellular engineering practice in general and engineered cell product manufacturing in particular.
Collapse
Affiliation(s)
- Harish G. Dixit
- Department of Bioengineering, University of California – Riverside, Riverside, California 92521, United States
| | - Renate Starr
- Department of Cancer Immunotherapy and Tumor Immunology, City of Hope Beckman Research Institute and Medical Center, Duarte, California 91010, United States
| | - Morgan L. Dundon
- Materials Science and Engineering Program, University of California – Riverside, Riverside, California 92521, United States
| | - Pranee I. Pairs
- Materials Science and Engineering Program, University of California – Riverside, Riverside, California 92521, United States
| | - Xin Yang
- Department of Cancer Immunotherapy and Tumor Immunology, City of Hope Beckman Research Institute and Medical Center, Duarte, California 91010, United States
| | - Yanyan Zhang
- Department of Mechanical Engineering, University of California – Riverside, Riverside, California 92521, United States
| | - Daniel Nampe
- Department of Bioengineering, University of California – Riverside, Riverside, California 92521, United States
- Department of Mechanical Engineering, University of California – Riverside, Riverside, California 92521, United States
| | - Christopher B. Ballas
- Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Hideaki Tsutsui
- Department of Bioengineering, University of California – Riverside, Riverside, California 92521, United States
- Department of Mechanical Engineering, University of California – Riverside, Riverside, California 92521, United States
- Stem Cell Center, University of California – Riverside, Riverside, California 92521, United States
| | - Stephen J. Forman
- Department of Cancer Immunotherapy and Tumor Immunology, City of Hope Beckman Research Institute and Medical Center, Duarte, California 91010, United States
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Beckman Research Institute and Medical Center, Duarte, California 91010, United States
| | - Christine E. Brown
- Department of Cancer Immunotherapy and Tumor Immunology, City of Hope Beckman Research Institute and Medical Center, Duarte, California 91010, United States
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Beckman Research Institute and Medical Center, Duarte, California 91010, United States
| | - Masaru P. Rao
- Department of Bioengineering, University of California – Riverside, Riverside, California 92521, United States
- Materials Science and Engineering Program, University of California – Riverside, Riverside, California 92521, United States
- Department of Mechanical Engineering, University of California – Riverside, Riverside, California 92521, United States
| |
Collapse
|
36
|
Guided migration analyses at the single-clone level uncover cellular targets of interest in tumor-associated myeloid-derived suppressor cell populations. Sci Rep 2020; 10:1189. [PMID: 31988310 PMCID: PMC6985212 DOI: 10.1038/s41598-020-57941-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 01/08/2020] [Indexed: 12/15/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are immune cells that exert immunosuppression within the tumor, protecting cancer cells from the host’s immune system and/or exogenous immunotherapies. While current research has been mostly focused in countering MDSC-driven immunosuppression, little is known about the mechanisms by which MDSCs disseminate/infiltrate cancerous tissue. This study looks into the use of microtextured surfaces, coupled with in vitro and in vivo cellular and molecular analysis tools, to videoscopically evaluate the dissemination patterns of MDSCs under structurally guided migration, at the single-cell level. MDSCs exhibited topographically driven migration, showing significant intra- and inter-population differences in motility, with velocities reaching ~40 μm h−1. Downstream analyses coupled with single-cell migration uncovered the presence of specific MDSC subpopulations with different degrees of tumor-infiltrating and anti-inflammatory capabilities. Granulocytic MDSCs showed a ~≥3-fold increase in maximum dissemination velocities and traveled distances, and a ~10-fold difference in the expression of pro- and anti-inflammatory markers. Prolonged culture also revealed that purified subpopulations of MDSCs exhibit remarkable plasticity, with homogeneous/sorted subpopulations giving rise to heterogenous cultures that represented the entire hierarchy of MDSC phenotypes within 7 days. These studies point towards the granulocytic subtype as a potential cellular target of interest given their superior dissemination ability and enhanced anti-inflammatory activity.
Collapse
|
37
|
Duarte-Sanmiguel S, Higuita-Castro N, Gallego-Perez D. Nanoelectroporation and Collection of Genetically Modified Exosomes in Primary Cultures of Dendritic Cells. Methods Mol Biol 2020; 2050:79-84. [PMID: 31468481 DOI: 10.1007/978-1-4939-9740-4_8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Dendritic cells (DCs) are cells of the immune system that behave as antigen presenters and assist in T cell activation. DCs have recently been used in cell-based immunotherapies for the treatment of different diseases due to the lack of adverse nonspecific immune responses, typically elicited by other approaches. Genetically modified DCs, for example, have been used to stimulate CD4/CD8 antigen presenting immune responses. However, genetic manipulation of primary DCs remains a challenge. Here we describe a protocol for nonviral, benign transfection of primary DCs using nanochannel-based electroporation, and the subsequent collection of genetically modified exosomes for downstream applications.
Collapse
Affiliation(s)
- Silvia Duarte-Sanmiguel
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA.,Department of Surgery, The Ohio State University, Columbus, OH, USA
| | - Natalia Higuita-Castro
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA.,Department of Surgery, The Ohio State University, Columbus, OH, USA
| | - Daniel Gallego-Perez
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA. .,Department of Surgery, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
38
|
Dong Z, Jiao Y, Xie B, Hao Y, Wang P, Liu Y, Shi J, Chitrakar C, Black S, Wang YC, Lee LJ, Li M, Fan Y, Chang L. On-chip multiplexed single-cell patterning and controllable intracellular delivery. MICROSYSTEMS & NANOENGINEERING 2020; 6:2. [PMID: 34567617 PMCID: PMC8433345 DOI: 10.1038/s41378-019-0112-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/23/2019] [Accepted: 10/10/2019] [Indexed: 05/03/2023]
Abstract
Conventional electroporation approaches show limitations in the delivery of macromolecules in vitro and in vivo. These limitations include low efficiency, noticeable cell damage and nonuniform delivery of cells. Here, we present a simple 3D electroporation platform that enables massively parallel single-cell manipulation and the intracellular delivery of macromolecules and small molecules. A pyramid pit micropore array chip was fabricated based on a silicon wet-etching method. A controllable vacuum system was adopted to trap a single cell on each micropore. Using this chip, safe single-cell electroporation was performed at low voltage. Cargoes of various sizes ranging from oligonucleotides (molecular beacons, 22 bp) to plasmid DNA (CRISPR-Cas9 expression vectors, >9 kb) were delivered into targeted cells with a significantly higher transfection efficiency than that of multiple benchmark methods (e.g., commercial electroporation devices and Lipofectamine). The delivered dose of the chemotherapeutic drug could be controlled by adjusting the applied voltage. By using CRISPR-Cas9 transfection with this system, the p62 gene and CXCR7 gene were knocked out in tumor cells, which effectively inhibited their cellular activity. Overall, this vacuum-assisted micropore array platform provides a simple, efficient, high-throughput intracellular delivery method that may facilitate on-chip cell manipulation, intracellular investigation and cancer therapy.
Collapse
Affiliation(s)
- Zaizai Dong
- School of Biological Science and Medical Engineering, Beihang University, 100083 Beijing, China
- Institute of Nanotechnology for Single Cell Analysis (INSCA), Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, 100083 Beijing, China
| | - Yanli Jiao
- College of Agricultural and Life Sciences, University of Florida, Gainesville, FL 32611 USA
| | - Bingteng Xie
- Center for Reproductive Medicine, Peking University Third Hospital, 100191 Beijing, China
| | - Yongcun Hao
- Ministry of Education Key Laboratory of Micro and Nano Systems for Aerospace, Northwestern Polytechnical University, 710072 Xi’an, China
| | - Pan Wang
- Center for Reproductive Medicine, Peking University Third Hospital, 100191 Beijing, China
| | - Yuanyuan Liu
- School of Biological Science and Medical Engineering, Beihang University, 100083 Beijing, China
- Institute of Nanotechnology for Single Cell Analysis (INSCA), Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, 100083 Beijing, China
| | - Junfeng Shi
- Chemical and Biomolecular Engineering Department, Ohio State University, Columbus, OH 43209 USA
| | - Chandani Chitrakar
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76207 USA
| | - Stephen Black
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76207 USA
| | - Yu-Chieh Wang
- Department of Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, TX 76107 USA
| | - L. James Lee
- Chemical and Biomolecular Engineering Department, Ohio State University, Columbus, OH 43209 USA
| | - Mo Li
- Center for Reproductive Medicine, Peking University Third Hospital, 100191 Beijing, China
| | - Yubo Fan
- School of Biological Science and Medical Engineering, Beihang University, 100083 Beijing, China
- Institute of Nanotechnology for Single Cell Analysis (INSCA), Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, 100083 Beijing, China
| | - Lingqian Chang
- School of Biological Science and Medical Engineering, Beihang University, 100083 Beijing, China
- Institute of Nanotechnology for Single Cell Analysis (INSCA), Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, 100083 Beijing, China
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76207 USA
| |
Collapse
|
39
|
Wu Y, Fu A, Yossifon G. Active particles as mobile microelectrodes for selective bacteria electroporation and transport. SCIENCE ADVANCES 2020; 6:eaay4412. [PMID: 32064350 PMCID: PMC6989140 DOI: 10.1126/sciadv.aay4412] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 11/22/2019] [Indexed: 05/16/2023]
Abstract
Self-propelling micromotors are emerging as a promising micro- and nanoscale tool for single-cell analysis. We have recently shown that the field gradients necessary to manipulate matter via dielectrophoresis can be induced at the surface of a polarizable active ("self-propelling") metallodielectric Janus particle (JP) under an externally applied electric field, acting essentially as a mobile floating microelectrode. Here, we successfully demonstrated that the application of an external electric field can singularly trap and transport bacteria and can selectively electroporate the trapped bacteria. Selective electroporation, enabled by the local intensification of the electric field induced by the JP, was obtained under both continuous alternating current and pulsed signal conditions. This approach is generic and applicable to bacteria and JP, as well as a wide range of cell types and micromotor designs. Hence, it constitutes an important and novel experimental tool for single-cell analysis and targeted delivery.
Collapse
Affiliation(s)
- Yue Wu
- Faculty of Mechanical Engineering, Micro- and Nanofluidics Laboratory, Technion–Israel Institute of Technology, Haifa 32000, Israel
| | - Afu Fu
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine and Research Institute, Technion–Israel Institute of Technology, Haifa 3109602, Israel
| | - Gilad Yossifon
- Faculty of Mechanical Engineering, Micro- and Nanofluidics Laboratory, Technion–Israel Institute of Technology, Haifa 32000, Israel
| |
Collapse
|
40
|
Tay A, Melosh N. Transfection with Nanostructure Electro‐Injection is Minimally Perturbative. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900133] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Andy Tay
- Department of Materials Science and Engineering Stanford University Stanford CA 94305 USA
- Department of Biomedical Engineering National University of Singapore Singapore 117583 Singapore
| | - Nicholas Melosh
- Department of Materials Science and Engineering Stanford University Stanford CA 94305 USA
| |
Collapse
|
41
|
Abstract
Intracellular cargo delivery is an essential step in many biomedical applications including gene editing and biologics therapy. Examples of cargo include nucleic acids (RNA and DNA), proteins, small biomolecules, and drugs, which can vary substantially in terms of their sizes, charges, solubility, and stability. Viruses have been used traditionally to deliver nucleic acids into cells, but the method suffers from limitations such as small cargo size, safety concerns, and viral genome integration into host cells, all of which complicate therapeutic applications. Commercially available techniques using biochemicals and bulk electroporation are, in general, poorly compatible with primary cells such as human induced pluripotent stem cells and immune cells, which are increasingly important candidates for adoptive cell therapy. Nanostructures, with dimensions ranging from tens of nanometers to a few micrometers, may play a critical role in overcoming cellular manipulation and delivery challenges and provide a powerful alternative to conventional techniques. A critical feature that differentiates nanostructures from viral, biochemical, and bulk electroporation techniques is that they interface with cells at a scale measuring ten to hundreds of nanometers in size. This highly local interaction enables application of stronger and more direct stimuli such as mechanical force, heat, or electric fields than would be possible in a bulk treatment. Compared to popular viral, biochemical, and bulk electroporation methods, nanostructures were found to minimally perturb cells with cells remaining in good health during postdelivery culture. These advantages have enabled nanostructures such as nanowires and nanotubes to successfully interface with a wide variety of cells, including primary immune cells and cardiomyocytes, for in vitro and in vivo applications. This Account is focused on using nanostructures for cargo delivery into biological cells. In this Account, we will first outline the historical developments using nanostructures for interfacing with cells. We will highlight how mechanistic understanding of nano-bio interactions has evolved over the last decade and how this improved knowledge has motivated coupling of electric and magnetic fields to nanostructures to improve delivery outcomes. There will also be an in-depth discussion on the merits of nanostructures in comparison to conventional methods using viruses, biochemicals, and bulk electroporation. Finally, motivated by our observations on the lack of consistency in reporting key metrics such as efficiency in literature, we suggest a set of metrics for documenting experimental results with the aim to promote standardization in reporting and ease in comparing. We suggest the use of more sophisticated tools such as RNA transcriptomics for thorough assessment of cell perturbation attributed to intracellular delivery. We hope that this Account can effectively capture the progress of nanostructure-mediated cargo delivery and encourage new innovations.
Collapse
Affiliation(s)
- Andy Tay
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States,Department of Biomedical Engineering, National University of Singapore, 117583 Singapore
| | - Nicholas Melosh
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States,Corresponding Author:
| |
Collapse
|
42
|
Soft Electroporation Through 3D Hollow Nanoelectrodes. Methods Mol Biol 2019. [PMID: 31468475 DOI: 10.1007/978-1-4939-9740-4_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Generally, electroporation of in vitro cells is performed under very high electric fields to overcome the physical barrier of plasma membrane. Since traditional electroporation techniques make use of very high voltages, which is critical to cell viability, this study presents a microfluidic platform able to perform cell membrane electroporation with the application of low voltages (1.5-2 V). The platform is manufactured based on the milling by mean of focused ionic beam, which offers an established approach to fabricate ordered arrays of 3D gold hollow nanoelectrodes protruding from an insulating substrate. The novelty of this fabrication relies on the fact that the nanoelectrodes used for electroporation are simultaneously metallic, hollow and communicate through its nanochannels with an isolated microfluidic chamber beneath the device. Adherent cultured cells on the nanoelectrodes can be electroporated in this platform, and molecules can be selectively delivered only inside the porated cells.
Collapse
|
43
|
3D Nanochannel Array for High-Throughput Cell Manipulation and Electroporation. Methods Mol Biol 2019. [PMID: 31468477 DOI: 10.1007/978-1-4939-9740-4_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Electroporation has been one of the most commonly used physical methods for gene/drug delivery. Compared to other nonviral counterparts, electroporation enables optimization of delivery efficiency by tuning the electric field applied on cells. Commercial electroporation, however, results in stochastic transfection and significant cellular damage mostly due to its "bulk" environment. In this chapter, we introduce nanoelectroporation (NEP) which has demonstrated living cell transfection in a highly controllable manner. In NEP, the electric field can be precisely focused on a single cell positioned on nanochannels. Safe single-cell electroporation as well as "electrophoretic" molecular delivery can be achieved on the same device. This system achieves significantly higher transfection efficiency and cellular viability than commercial systems. This device is unique in that it can efficiently deliver genetic molecules (e.g., DNAs, RNAs) that exceed 10 kbp in size. The NEP device based on a 3D nanochannel array prototype was fabricated using cleanroom techniques. For achieving precise cell to nanochannel pairing, three on-chip high-throughput manipulation technologies were developed, that is, magnetic tweezers (MT), dielectrophoresis (DEP), and thin-film microfluidics.
Collapse
|
44
|
Punjiya M, Nejad HR, Mathews J, Levin M, Sonkusale S. A flow through device for simultaneous dielectrophoretic cell trapping and AC electroporation. Sci Rep 2019; 9:11988. [PMID: 31427614 PMCID: PMC6700080 DOI: 10.1038/s41598-019-48198-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 07/30/2019] [Indexed: 02/06/2023] Open
Abstract
Isolation of cells and their transfection in a controlled manner is an integral step in cell biotechnology. Electric field approaches such as dielectrophoresis (DEP) offers a more viable method for targeted immobilization of cells without any labels. For transfection of cells to incorporate exogenous materials, electrical methods such as electroporation, are preferred over chemical and viral delivery methods since they minimally affect cell viability and can target many types. However prior approaches to both methods required multiple excitation sources, an AC source for DEP-based trapping and another DC source for electroporation. In this paper, we present a first of its kind flow through lab-on-chip platform using a single AC excitation source for combined trapping using negative dielectrophoresis (nDEP) and AC electroporation. Use of AC fields for electroporation eliminates the unwanted side effects of electrolysis or joule heating at electrodes compared to DC electroporation. Adjusting the flow rate and the electrical parameters of the incident AC field precisely controls the operation (trap, trap with electroporation and release). The platform has been validated through trapping and simultaneous transfection of HEK-293 embryonic kidney cells with a plasmid vector containing a fluorescent protein tag. Numerical scaling analysis is provided that indicates promise for individual cell trapping and electroporation using low voltage AC fields.
Collapse
Affiliation(s)
- Meera Punjiya
- Tufts University, Department of Electrical and Computer Engineering, 161 College Ave, Medford, MA, 02155, USA.,Nano Lab, Advanced Technology Laboratory, 200 Boston Ave, Medford, MA, 02155, USA
| | - Hojatollah Rezaei Nejad
- Tufts University, Department of Electrical and Computer Engineering, 161 College Ave, Medford, MA, 02155, USA.,Nano Lab, Advanced Technology Laboratory, 200 Boston Ave, Medford, MA, 02155, USA
| | - Juanita Mathews
- Allen Discovery Center at Tufts University, Department of Biology, Medford, MA, 02155, USA
| | - Michael Levin
- Allen Discovery Center at Tufts University, Department of Biology, Medford, MA, 02155, USA
| | - Sameer Sonkusale
- Tufts University, Department of Electrical and Computer Engineering, 161 College Ave, Medford, MA, 02155, USA. .,Nano Lab, Advanced Technology Laboratory, 200 Boston Ave, Medford, MA, 02155, USA.
| |
Collapse
|
45
|
Abstract
Nanostructured devices are able to foster the technology for cell membrane poration. With the size smaller than a cell, nanostructures allow efficient poration on the cell membrane. Emerging nanostructures with various physical transduction have been demonstrated to accommodate effective intracellular delivery. Aside from improving poration and intracellular delivery performance, nanostructured devices also allow for the discovery of novel physiochemical phenomena and the biological response of the cell. This article provides a brief introduction to the principles of nanostructured devices for cell poration and outlines the intracellular delivery capability of the technology. In the future, we envision more exploration on new nanostructure designs and creative applications in biomedical fields.
Collapse
Affiliation(s)
- Apresio K Fajrial
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, 80309 United States of America
| | | |
Collapse
|
46
|
Shi J, Ma Y, Zhu J, Chen Y, Sun Y, Yao Y, Yang Z, Xie J. A Review on Electroporation-Based Intracellular Delivery. Molecules 2018; 23:E3044. [PMID: 30469344 PMCID: PMC6278265 DOI: 10.3390/molecules23113044] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 11/13/2018] [Accepted: 11/17/2018] [Indexed: 12/17/2022] Open
Abstract
Intracellular delivery is a critical step in biological discoveries and has been widely utilized in biomedical research. A variety of molecular tools have been developed for cell-based gene therapies, including FDA approved CAR-T immunotherapy, iPSC, cell reprogramming and gene editing. Despite the inspiring results of these applications, intracellular delivery of foreign molecules including nucleic acids and proteins remains challenging. Efficient yet non-invasive delivery of biomolecules in a high-throughput manner has thus long fascinates the scientific community. As one of the most popular non-viral technologies for cell transfection, electroporation has gone through enormous development with the assist of nanotechnology and microfabrication. Emergence of miniatured electroporation system brought up many merits over the weakness of traditional electroporation system, including precise dose control and high cell viability. These new generation of electroporation systems are of considerable importance to expand the biological applications of intracellular delivery, bypassing the potential safety issue of viral vectors. In this review, we will go over the recent progresses in the electroporation-based intracellular delivery and several potential applications of cutting-edge research on the miniatured electroporation, including gene therapy, cellular reprogramming and intracellular probe.
Collapse
Affiliation(s)
- Junfeng Shi
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA.
| | - Yifan Ma
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA.
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA.
| | - Jing Zhu
- College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA.
| | - Yuanxin Chen
- Department of Neurosurgery, Mayo Clinic College of Medicine, Jacksonville, FL 33573, USA.
| | - Yating Sun
- School of Life Sciences, Jilin University, Changchun 130012, China.
| | - Yicheng Yao
- School of Life Sciences, Jilin University, Changchun 130012, China.
| | - Zhaogang Yang
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA.
| | - Jing Xie
- School of Life Sciences, Jilin University, Changchun 130012, China.
| |
Collapse
|
47
|
Ansón-Casaos A, Grasa L, Pereboom D, Mesonero JE, Casanova A, Murillo MD, Martínez MT. In-vitro toxicity of carbon nanotube/polylysine colloids to colon cancer cells. IET Nanobiotechnol 2018; 10:374-381. [PMID: 27906137 DOI: 10.1049/iet-nbt.2015.0088] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Single-walled carbon nanotubes (SWCNTs) are thoroughly purified and dispersed in an aqueous solution of high molecular weight poly-L-lysine (pLlys). Human intestinal epithelial Caco-2/TC7 cells are incubated with the SWCNT dispersions in pLlys, and their effects on cell viability are studied by image flow cytometry. No significant changes are observed in the cell culture wells up to pLlys concentrations of 10 μg ml-1. However, high mortality is detected at pLlys concentrations of 100 μg ml-1. The presence of oxygen-free SWCNTs does not modify the effects of pLlys on cell cultures at any of the tested concentrations (≤1 μg ml-1). In addition, SWCNTs having an 8 wt.% of surface oxygen are tested with identical results. Thus, purified SWCNTs, even bearing oxygen functional groups, act as inert particles in the cell culture medium. This result supports the applicability of SWCNTs as carriers in pharmacological formulations against digestive tract diseases.
Collapse
Affiliation(s)
| | - Laura Grasa
- Instituto Agroalimentario de Aragón (IA2) (Universidad de Zaragoza - CITA), Zaragoza, Spain
| | - Desirée Pereboom
- Servicio de Citómica, Universidad de Zaragoza, Domingo Miral s/n, 50009 Zaragoza, Spain
| | - José Emilio Mesonero
- Instituto Agroalimentario de Aragón (IA2) (Universidad de Zaragoza - CITA), Zaragoza, Spain
| | - Alvaro Casanova
- Departamento de Farmacología y Fisiología, Facultad de Medicina, Universidad de Zaragoza, Domingo Miral s/n, 50009 Zaragoza, Spain
| | - María Divina Murillo
- Departamento de Farmacología y Fisiología, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón (IIS), Universidad de Zaragoza, Miguel Servet 177, 50013 Zaragoza, Spain
| | | |
Collapse
|
48
|
Wu H, Zhu J, Huang Y, Wu D, Sun J. Microfluidic-Based Single-Cell Study: Current Status and Future Perspective. Molecules 2018; 23:E2347. [PMID: 30217082 PMCID: PMC6225124 DOI: 10.3390/molecules23092347] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 09/05/2018] [Accepted: 09/09/2018] [Indexed: 01/29/2023] Open
Abstract
Investigation of cell behavior under different environments and manual operations can give information in specific cellular processes. Among all cell-based analysis, single-cell study occupies a peculiar position, while it can avoid the interaction effect within cell groups and provide more precise information. Microfluidic devices have played an increasingly important role in the field of single-cell study owing to their advantages: high efficiency, easy operation, and low cost. In this review, the applications of polymer-based microfluidics on cell manipulation, cell treatment, and cell analysis at single-cell level are detailed summarized. Moreover, three mainly types of manufacturing methods, i.e., replication, photodefining, and soft lithography methods for polymer-based microfluidics are also discussed.
Collapse
Affiliation(s)
- Haiwa Wu
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
| | - Jing Zhu
- Department of Pharmaceutics, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA.
| | - Yao Huang
- College of Mechanical and Electrical Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Daming Wu
- College of Mechanical and Electrical Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
- State Key Laboratory of Organic-Inorganic Composites, Beijing 100029, China.
| | - Jingyao Sun
- College of Mechanical and Electrical Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
49
|
Hong J, Kim B, Shin H. Mixed-scale poly(methyl methacrylate) channel network-based single-particle manipulation via diffusiophoresis. NANOSCALE 2018; 10:14421-14431. [PMID: 29796559 DOI: 10.1039/c7nr07669j] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Despite the unique advantages of nanochannels imparted by their small size, their utility is limited by the lack of affordable and versatile fabrication methods. Moreover, nanochannel-incorporated fluidic devices require micro-sized conduit integration for efficient access of liquid samples. In this study, a simple and cost-effective fabrication method for mixed-scale channel networks via hot-embossing of poly(methyl methacrylate) (PMMA) using a carbon stamp is demonstrated. Due to its high rigidity, PMMA ensures collapse-free channel fabrication. The carbon stamp is fabricated using only batch microfabrication and has a convex architecture that allows the fabrication of a complex channel network via a single imprinting process. In addition, the microchannels are connected to nanochannels via three-dimensional (3D) microfunnels that serve as single-particle-entrapment chambers, ensuring smooth transport of samples into the nanochannels. Owing to the 3D geometry of the microfunnels and the small size of the nanochannels, a solute gradient can be generated locally at the microfunnel. This local solute gradient enables the entrapment of microparticles at the microfunnels via diffusiophoresis, which can manipulate the particle motion in a controllable manner, without any external equipment or additional electrode integration into the channels. To the best of our knowledge, this is the first report of diffusiophoresis-based single-particle entrapment.
Collapse
Affiliation(s)
- Jisoo Hong
- Department of Mechanical Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea.
| | | | | |
Collapse
|
50
|
Shukla VC, Kuang TR, Senthilvelan A, Higuita-Castro N, Duarte-Sanmiguel S, Ghadiali SN, Gallego-Perez D. Lab-on-a-Chip Platforms for Biophysical Studies of Cancer with Single-Cell Resolution. Trends Biotechnol 2018; 36:549-561. [PMID: 29559164 DOI: 10.1016/j.tibtech.2018.02.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 02/15/2018] [Accepted: 02/16/2018] [Indexed: 12/14/2022]
Abstract
Recent cancer research has more strongly emphasized the biophysical aspects of tumor development, progression, and microenvironment. In addition to genetic modifications and mutations in cancer cells, it is now well accepted that the physical properties of cancer cells such as stiffness, electrical impedance, and refractive index vary with tumor progression and can identify a malignant phenotype. Moreover, cancer heterogeneity renders population-based characterization techniques inadequate, as individual cellular features are lost in the average. Hence, platforms for fast and accurate characterization of biophysical properties of cancer cells at the single-cell level are required. Here, we highlight some of the recent advances in the field of cancer biophysics and the development of lab-on-a-chip platforms for single-cell biophysical analyses of cancer cells.
Collapse
Affiliation(s)
- Vasudha C Shukla
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine and Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA; Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH 43210, USA; These authors contributed equally to this work
| | - Tai-Rong Kuang
- The Key Laboratory of Polymer Processing Engineering of Ministry of Education, South China University of Technology, Guangzhou 510640, P.R. China; These authors contributed equally to this work.
| | - Abirami Senthilvelan
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Natalia Higuita-Castro
- Department of Internal Medicine (Division of Pulmonary, Critical Care and Sleep Medicine), Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA; Department of Surgery, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Silvia Duarte-Sanmiguel
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH 43210, USA; Department of Human Sciences (Human Nutrition), College of Human Ecology, The Ohio State University, Columbus, OH 43210, USA
| | - Samir N Ghadiali
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH 43210, USA; Department of Internal Medicine (Division of Pulmonary, Critical Care and Sleep Medicine), Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Daniel Gallego-Perez
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH 43210, USA; Department of Surgery, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|