1
|
Lai Y, Tao W, Wang L, Liu Z, Wu P, Yang G, Yuan L. Medical Ultrasound Application Beyond Diagnosis: Insights From Ultrasound Sensing and Biological Response. Biotechnol J 2024; 19:e202400561. [PMID: 39726053 DOI: 10.1002/biot.202400561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/09/2024] [Accepted: 11/30/2024] [Indexed: 12/28/2024]
Abstract
Ultrasound (US) can easily penetrate media with excellent spatial precision corresponding to its wavelength. Naturally, US plays a pivotal role in the echolocation abilities of certain mammals such as bats and dolphins. In addition, medical US generated by transducers interact with tissues via delivering ultrasonic energy in the modes of heat generation, exertion of acoustic radiation force (ARF), and acoustic cavitation. Based on the principle of echolocation, various assistive devices for visual impairment people have been developed. High-Intensity Focused Ultrasound (HIFU) are developed for targeted ablation and tissue destruction. Besides thermal ablation, histotripsy with US is designed to damage tissue purely via mechanical effect without thermal coagulation. Low-Intensity Focused Ultrasound (LIFU) has been proven to be an effective stimulation method for neuromodulation. Furthermore, US has been reported to transiently increase the permeability of biological membranes, enabling acoustic transfection and blood-brain barrier open. All of these advances in US are changing the clinic. This review mainly introduces the advances in these aspects, focusing on the physical and biological principles, challenges, and future direction.
Collapse
Affiliation(s)
- Yubo Lai
- Department of Ultrasound Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Wenxin Tao
- Department of Ultrasound Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Lantian Wang
- Department of Ultrasound Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Zhaoyou Liu
- Department of Ultrasound Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Pengying Wu
- Department of Ultrasound Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Guodong Yang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University Xi'an, Xi'an, Shaanxi, China
| | - Lijun Yuan
- Department of Ultrasound Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
2
|
Linke JA, Munn LL, Jain RK. Compressive stresses in cancer: characterization and implications for tumour progression and treatment. Nat Rev Cancer 2024; 24:768-791. [PMID: 39390249 DOI: 10.1038/s41568-024-00745-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/20/2024] [Indexed: 10/12/2024]
Abstract
Beyond their many well-established biological aberrations, solid tumours create an abnormal physical microenvironment that fuels cancer progression and confers treatment resistance. Mechanical forces impact tumours across a range of biological sizes and timescales, from rapid events at the molecular level involved in their sensing and transmission, to slower and larger-scale events, including clonal selection, epigenetic changes, cell invasion, metastasis and immune response. Owing to challenges with studying these dynamic stimuli in biological systems, the mechanistic understanding of the effects and pathways triggered by abnormally elevated mechanical forces remains elusive, despite clear correlations with cancer pathophysiology, aggressiveness and therapeutic resistance. In this Review, we examine the emerging and diverse roles of physical forces in solid tumours and provide a comprehensive framework for understanding solid stress mechanobiology. We first review the physiological importance of mechanical forces, especially compressive stresses, and discuss their defining characteristics, biological context and relative magnitudes. We then explain how abnormal compressive stresses emerge in tumours and describe the experimental challenges in investigating these mechanically induced processes. Finally, we discuss the clinical translation of mechanotherapeutics that alleviate solid stresses and their potential to synergize with chemotherapy, radiotherapy and immunotherapies.
Collapse
Affiliation(s)
- Julia A Linke
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Lance L Munn
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
3
|
Li Y, Li XM, Wei LS, Ye JF. Advancements in mitochondrial-targeted nanotherapeutics: overcoming biological obstacles and optimizing drug delivery. Front Immunol 2024; 15:1451989. [PMID: 39483479 PMCID: PMC11524880 DOI: 10.3389/fimmu.2024.1451989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 09/19/2024] [Indexed: 11/03/2024] Open
Abstract
In recent decades, nanotechnology has significantly advanced drug delivery systems, particularly in targeting subcellular organelles, thus opening new avenues for disease treatment. Mitochondria, critical for cellular energy and health, when dysfunctional, contribute to cancer, neurodegenerative diseases, and metabolic disorders. This has propelled the development of nanomedicines aimed at precise mitochondrial targeting to modulate their function, marking a research hotspot. This review delves into the recent advancements in mitochondrial-targeted nanotherapeutics, with a comprehensive focus on targeting strategies, nanocarrier designs, and their therapeutic applications. It emphasizes nanotechnology's role in enhancing drug delivery by overcoming biological barriers and optimizing drug design for specific mitochondrial targeting. Strategies exploiting mitochondrial membrane potential differences and specific targeting ligands improve the delivery and mitochondrial accumulation of nanomedicines. The use of diverse nanocarriers, including liposomes, polymer nanoparticles, and inorganic nanoparticles, tailored for effective mitochondrial targeting, shows promise in anti-tumor and neurodegenerative treatments. The review addresses the challenges and future directions in mitochondrial targeting nanotherapy, highlighting the need for precision, reduced toxicity, and clinical validation. Mitochondrial targeting nanotherapy stands at the forefront of therapeutic strategies, offering innovative treatment perspectives. Ongoing innovation and research are crucial for developing more precise and effective treatment modalities.
Collapse
Affiliation(s)
- Yang Li
- General Surgery Center, First Hospital of Jilin University, Changchun, China
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Xiao-meng Li
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Li-si Wei
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Jun-feng Ye
- General Surgery Center, First Hospital of Jilin University, Changchun, China
| |
Collapse
|
4
|
Kim MG, Yoon C, Lim HG. Recent Advancements in High-Frequency Ultrasound Applications from Imaging to Microbeam Stimulation. SENSORS (BASEL, SWITZERLAND) 2024; 24:6471. [PMID: 39409511 PMCID: PMC11479296 DOI: 10.3390/s24196471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/18/2024] [Accepted: 10/03/2024] [Indexed: 10/20/2024]
Abstract
Ultrasound is a versatile and well-established technique using sound waves with frequencies higher than the upper limit of human hearing. Typically, therapeutic and diagnosis ultrasound operate in the frequency range of 500 kHz to 15 MHz with greater depth of penetration into the body. However, to achieve improved spatial resolution, high-frequency ultrasound (>15 MHz) was recently introduced and has shown promise in various fields such as high-resolution imaging for the morphological features of the eye and skin as well as small animal imaging for drug and gene therapy. In addition, high-frequency ultrasound microbeam stimulation has been demonstrated to manipulate single cells or microparticles for the elucidation of physical and functional characteristics of cells with minimal effect on normal cell physiology and activity. Furthermore, integrating machine learning with high-frequency ultrasound enhances diagnostics, including cell classification, cell deformability estimation, and the diagnosis of diabetes and dysnatremia using convolutional neural networks (CNNs). In this paper, current efforts in the use of high-frequency ultrasound from imaging to stimulation as well as the integration of deep learning are reviewed, and potential biomedical and cellular applications are discussed.
Collapse
Affiliation(s)
- Min Gon Kim
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90007, USA
| | - Changhan Yoon
- Department of Biomedical Engineering, Inje University, Gimhae 50834, Republic of Korea
| | - Hae Gyun Lim
- Department of Biomedical Engineering, Pukyong National University, Busan 48547, Republic of Korea;
| |
Collapse
|
5
|
Huang G, Lin L, Liu Q, Wu S, Chen J, Zhu R, You H, Sun C. Three-dimensional array of microbubbles sonoporation of cells in microfluidics. Front Bioeng Biotechnol 2024; 12:1353333. [PMID: 38419723 PMCID: PMC10899490 DOI: 10.3389/fbioe.2024.1353333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 01/29/2024] [Indexed: 03/02/2024] Open
Abstract
Sonoporation is a popular membrane disruption technique widely applicable in various fields, including cell therapy, drug delivery, and biomanufacturing. In recent years, there has been significant progress in achieving controlled, high-viability, and high-efficiency cell sonoporation in microfluidics. If the microchannels are too small, especially when scaled down to the cellular level, it still remains a challenge to overcome microchannel clogging, and low throughput. Here, we presented a microfluidic device capable of modulating membrane permeability through oscillating three-dimensional array of microbubbles. Simulations were performed to analyze the effective range of action of the oscillating microbubbles to obtain the optimal microchannel size. Utilizing a high-precision light curing 3D printer to fabricate uniformly sized microstructures in a one-step on both the side walls and the top surface for the generation of microbubbles. These microbubbles oscillated with nearly identical amplitudes and frequencies, ensuring efficient and stable sonoporation within the system. Cells were captured and trapped on the bubble surface by the acoustic streaming and secondary acoustic radiation forces induced by the oscillating microbubbles. At a driving voltage of 30 Vpp, the sonoporation efficiency of cells reached 93.9% ± 2.4%.
Collapse
Affiliation(s)
- Guangyong Huang
- School of Mechanical Engineering, Guangxi University, Nanning, China
- School of Mechanical and Automotive Engineering, Guangxi University of Science and Technology, Liuzhou, China
| | - Lin Lin
- School of Mechanical Engineering, Guangxi University, Nanning, China
| | - Quanhui Liu
- Animal Science and Technology College, Guangxi University, Nanning, China
| | - Shixiong Wu
- School of Mechanical Engineering, Guangxi University, Nanning, China
| | - Jiapeng Chen
- School of Mechanical Engineering, Guangxi University, Nanning, China
| | - Rongxing Zhu
- School of Mechanical Engineering, Guangxi University, Nanning, China
| | - Hui You
- School of Mechanical Engineering, Guangxi University, Nanning, China
| | - Cuimin Sun
- School of Computer, Electronics and Information, Guangxi University, Nanning, China
| |
Collapse
|
6
|
Shokouhi AR, Chen Y, Yoh HZ, Murayama T, Suu K, Morikawa Y, Brenker J, Alan T, Voelcker NH, Elnathan R. Electroactive nanoinjection platform for intracellular delivery and gene silencing. J Nanobiotechnology 2023; 21:273. [PMID: 37592297 PMCID: PMC10433684 DOI: 10.1186/s12951-023-02056-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 08/07/2023] [Indexed: 08/19/2023] Open
Abstract
BACKGROUND Nanoinjection-the process of intracellular delivery using vertically configured nanostructures-is a physical route that efficiently negotiates the plasma membrane, with minimal perturbation and toxicity to the cells. Nanoinjection, as a physical membrane-disruption-mediated approach, overcomes challenges associated with conventional carrier-mediated approaches such as safety issues (with viral carriers), genotoxicity, limited packaging capacity, low levels of endosomal escape, and poor versatility for cell and cargo types. Yet, despite the implementation of nanoinjection tools and their assisted analogues in diverse cellular manipulations, there are still substantial challenges in harnessing these platforms to gain access into cell interiors with much greater precision without damaging the cell's intricate structure. Here, we propose a non-viral, low-voltage, and reusable electroactive nanoinjection (ENI) platform based on vertically configured conductive nanotubes (NTs) that allows for rapid influx of targeted biomolecular cargos into the intracellular environment, and for successful gene silencing. The localization of electric fields at the tight interface between conductive NTs and the cell membrane drastically lowers the voltage required for cargo delivery into the cells, from kilovolts (for bulk electroporation) to only ≤ 10 V; this enhances the fine control over membrane disruption and mitigates the problem of high cell mortality experienced by conventional electroporation. RESULTS Through both theoretical simulations and experiments, we demonstrate the capability of the ENI platform to locally perforate GPE-86 mouse fibroblast cells and efficiently inject a diverse range of membrane-impermeable biomolecules with efficacy of 62.5% (antibody), 55.5% (mRNA), and 51.8% (plasmid DNA), with minimal impact on cells' viability post nanoscale-EP (> 90%). We also show gene silencing through the delivery of siRNA that targets TRIOBP, yielding gene knockdown efficiency of 41.3%. CONCLUSIONS We anticipate that our non-viral and low-voltage ENI platform is set to offer a new safe path to intracellular delivery with broader selection of cargo and cell types, and will open opportunities for advanced ex vivo cell engineering and gene silencing.
Collapse
Affiliation(s)
- Ali-Reza Shokouhi
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
| | - Yaping Chen
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
| | - Hao Zhe Yoh
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
| | - Takahide Murayama
- Institute of Semiconductor and Electronics Technologies, ULVAC Inc, 1220-1 Suyama, Susono, Shizuoka, 410-1231, Japan
| | - Koukou Suu
- Institute of Semiconductor and Electronics Technologies, ULVAC Inc, 1220-1 Suyama, Susono, Shizuoka, 410-1231, Japan
| | - Yasuhiro Morikawa
- Institute of Semiconductor and Electronics Technologies, ULVAC Inc, 1220-1 Suyama, Susono, Shizuoka, 410-1231, Japan
| | - Jason Brenker
- Department of Mechanical and Aerospace Engineering, Monash University, Wellington Rd, Clayton, VIC, 3168, Australia
| | - Tuncay Alan
- Department of Mechanical and Aerospace Engineering, Monash University, Wellington Rd, Clayton, VIC, 3168, Australia
| | - Nicolas H Voelcker
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia.
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia.
- INM-Leibniz Institute for New Materials, Campus D2 2, 66123, Saarbrücken, Germany.
- Department of Materials Science and Engineering, Monash University, 22 Alliance Lane, Clayton, VIC, 3168, Australia.
| | - Roey Elnathan
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia.
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia.
- Faculty of Health, School of Medicine, Deakin University, Waurn Ponds, Melbourne, VIC, 3216, Australia.
- Institute for Frontier Materials, Deakin University, Geelong Waurn Ponds campus, Melbourne, VIC, 3216, Australia.
- The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong Waurn Ponds Campus, Melbourne, VIC, 3216, Australia.
| |
Collapse
|
7
|
Isenmann M, Stoddart MJ, Schmelzeisen R, Gross C, Della Bella E, Rothweiler RM. Basic Principles of RNA Interference: Nucleic Acid Types and In Vitro Intracellular Delivery Methods. MICROMACHINES 2023; 14:1321. [PMID: 37512632 PMCID: PMC10383872 DOI: 10.3390/mi14071321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023]
Abstract
Since its discovery in 1989, RNA interference (RNAi) has become a widely used tool for the in vitro downregulation of specific gene expression in molecular biological research. This basically involves a complementary RNA that binds a target sequence to affect its transcription or translation process. Currently, various small RNAs, such as small interfering RNA (siRNA), micro RNA (miRNA), small hairpin RNA (shRNA), and PIWI interacting RNA (piRNA), are available for application on in vitro cell culture, to regulate the cells' gene expression by mimicking the endogenous RNAi-machinery. In addition, several biochemical, physical, and viral methods have been established to deliver these RNAs into the cell or nucleus. Since each RNA and each delivery method entail different off-target effects, limitations, and compatibilities, it is crucial to understand their basic mode of action. This review is intended to provide an overview of different nucleic acids and delivery methods for planning, interpreting, and troubleshooting of RNAi experiments.
Collapse
Affiliation(s)
- Marie Isenmann
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, University of Freiburg, Hugstetterstrasse 55, 79106 Freiburg, Germany
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland
| | - Martin James Stoddart
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, University of Freiburg, Hugstetterstrasse 55, 79106 Freiburg, Germany
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland
| | - Rainer Schmelzeisen
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, University of Freiburg, Hugstetterstrasse 55, 79106 Freiburg, Germany
| | - Christian Gross
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, University of Freiburg, Hugstetterstrasse 55, 79106 Freiburg, Germany
| | - Elena Della Bella
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland
| | - René Marcel Rothweiler
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, University of Freiburg, Hugstetterstrasse 55, 79106 Freiburg, Germany
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland
| |
Collapse
|
8
|
Campelo SN, Huang PH, Buie CR, Davalos RV. Recent Advancements in Electroporation Technologies: From Bench to Clinic. Annu Rev Biomed Eng 2023; 25:77-100. [PMID: 36854260 PMCID: PMC11633374 DOI: 10.1146/annurev-bioeng-110220-023800] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
Over the past decade, the increased adoption of electroporation-based technologies has led to an expansion of clinical research initiatives. Electroporation has been utilized in molecular biology for mammalian and bacterial transfection; for food sanitation; and in therapeutic settings to increase drug uptake, for gene therapy, and to eliminate cancerous tissues. We begin this article by discussing the biophysics required for understanding the concepts behind the cell permeation phenomenon that is electroporation. We then review nano- and microscale single-cell electroporation technologies before scaling up to emerging in vivo applications.
Collapse
Affiliation(s)
- Sabrina N Campelo
- Department of Biomedical Engineering and Mechanics, Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Virginia Tech, Blacksburg, Virginia, USA;
| | - Po-Hsun Huang
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Cullen R Buie
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Rafael V Davalos
- Department of Biomedical Engineering and Mechanics, Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Virginia Tech, Blacksburg, Virginia, USA;
| |
Collapse
|
9
|
Ambattu LA, Yeo LY. Sonomechanobiology: Vibrational stimulation of cells and its therapeutic implications. BIOPHYSICS REVIEWS 2023; 4:021301. [PMID: 38504927 PMCID: PMC10903386 DOI: 10.1063/5.0127122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 02/27/2023] [Indexed: 03/21/2024]
Abstract
All cells possess an innate ability to respond to a range of mechanical stimuli through their complex internal machinery. This comprises various mechanosensory elements that detect these mechanical cues and diverse cytoskeletal structures that transmit the force to different parts of the cell, where they are transcribed into complex transcriptomic and signaling events that determine their response and fate. In contrast to static (or steady) mechanostimuli primarily involving constant-force loading such as compression, tension, and shear (or forces applied at very low oscillatory frequencies (≤ 1 Hz) that essentially render their effects quasi-static), dynamic mechanostimuli comprising more complex vibrational forms (e.g., time-dependent, i.e., periodic, forcing) at higher frequencies are less well understood in comparison. We review the mechanotransductive processes associated with such acoustic forcing, typically at ultrasonic frequencies (> 20 kHz), and discuss the various applications that arise from the cellular responses that are generated, particularly for regenerative therapeutics, such as exosome biogenesis, stem cell differentiation, and endothelial barrier modulation. Finally, we offer perspectives on the possible existence of a universal mechanism that is common across all forms of acoustically driven mechanostimuli that underscores the central role of the cell membrane as the key effector, and calcium as the dominant second messenger, in the mechanotransduction process.
Collapse
Affiliation(s)
- Lizebona August Ambattu
- Micro/Nanophysics Research Laboratory, School of Engineering, RMIT University, Melbourne VIC 3000, Australia
| | - Leslie Y. Yeo
- Micro/Nanophysics Research Laboratory, School of Engineering, RMIT University, Melbourne VIC 3000, Australia
| |
Collapse
|
10
|
Figarol A, Olive L, Joubert O, Ferrari L, Rihn BH, Sarry F, Beyssen D. Biological Effects and Applications of Bulk and Surface Acoustic Waves on In Vitro Cultured Mammal Cells: New Insights. Biomedicines 2022; 10:biomedicines10051166. [PMID: 35625902 PMCID: PMC9139135 DOI: 10.3390/biomedicines10051166] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/09/2022] [Accepted: 05/13/2022] [Indexed: 02/01/2023] Open
Abstract
Medical imaging has relied on ultrasound (US) as an exploratory method for decades. Nonetheless, in cell biology, the numerous US applications are mainly in the research and development phase. In this review, we report the main effects on human or mammal cells of US induced by bulk or surface acoustic waves (SAW). At low frequencies, bulk US can lead to cell death. Under specific intensities and exposure times, however, cell proliferation and migration can be enhanced through cytoskeleton fluidization (a reorganization of the actin filaments and microtubules). Cavitation phenomena, frequencies of resonance close to those of the biological compounds, and mechanical transfers of energy from the acoustic pressure could explain those biological outcomes. At higher frequencies, no cavitation is observed. However, USs of high frequency stimulate ionic channels and increase cell permeability and transfection potency. Surface acoustic waves are increasingly exploited in microfluidics, especially for precise cell manipulations and cell sorting. With applications in diagnosis, infection, cancer treatment, or wound healing, US has remarkable potential. More mechanotransduction studies would be beneficial to understand the distinct roles of temperature rise, acoustic streaming and mechanical and electrical stimuli in the field.
Collapse
Affiliation(s)
- Agathe Figarol
- Institut FEMTO-ST, UMR CNRS 6174, Université de Bourgogne Franche-Comté, F-25030 Besançon, France;
- Institut Jean Lamour, UMR CNRS 7198, Université de Lorraine, CNRS, IJL, F-54000 Nancy, France; (L.O.); (O.J.); (L.F.); (B.H.R.); (F.S.)
| | - Lucile Olive
- Institut Jean Lamour, UMR CNRS 7198, Université de Lorraine, CNRS, IJL, F-54000 Nancy, France; (L.O.); (O.J.); (L.F.); (B.H.R.); (F.S.)
| | - Olivier Joubert
- Institut Jean Lamour, UMR CNRS 7198, Université de Lorraine, CNRS, IJL, F-54000 Nancy, France; (L.O.); (O.J.); (L.F.); (B.H.R.); (F.S.)
| | - Luc Ferrari
- Institut Jean Lamour, UMR CNRS 7198, Université de Lorraine, CNRS, IJL, F-54000 Nancy, France; (L.O.); (O.J.); (L.F.); (B.H.R.); (F.S.)
| | - Bertrand H. Rihn
- Institut Jean Lamour, UMR CNRS 7198, Université de Lorraine, CNRS, IJL, F-54000 Nancy, France; (L.O.); (O.J.); (L.F.); (B.H.R.); (F.S.)
| | - Frédéric Sarry
- Institut Jean Lamour, UMR CNRS 7198, Université de Lorraine, CNRS, IJL, F-54000 Nancy, France; (L.O.); (O.J.); (L.F.); (B.H.R.); (F.S.)
| | - Denis Beyssen
- Institut Jean Lamour, UMR CNRS 7198, Université de Lorraine, CNRS, IJL, F-54000 Nancy, France; (L.O.); (O.J.); (L.F.); (B.H.R.); (F.S.)
- Correspondence: ; Tel.: +33-61-448-6182
| |
Collapse
|
11
|
Li J, Ma Y, Zhang T, Shung KK, Zhu B. Recent Advancements in Ultrasound Transducer: From Material Strategies to Biomedical Applications. BME FRONTIERS 2022; 2022:9764501. [PMID: 37850168 PMCID: PMC10521713 DOI: 10.34133/2022/9764501] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 02/06/2022] [Indexed: 10/19/2023] Open
Abstract
Ultrasound is extensively studied for biomedical engineering applications. As the core part of the ultrasonic system, the ultrasound transducer plays a significant role. For the purpose of meeting the requirement of precision medicine, the main challenge for the development of ultrasound transducer is to further enhance its performance. In this article, an overview of recent developments in ultrasound transducer technologies that use a variety of material strategies and device designs based on both the piezoelectric and photoacoustic mechanisms is provided. Practical applications are also presented, including ultrasound imaging, ultrasound therapy, particle/cell manipulation, drug delivery, and nerve stimulation. Finally, perspectives and opportunities are also highlighted.
Collapse
Affiliation(s)
- Jiapu Li
- Wuhan National Laboratory for Optoelectronics, Optics Valley Laboratory, School of Optical and Electronic Information, Huazhong University of Science and Technology, Wuhan, China, 430074
- State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai 200050, China
| | - Yuqing Ma
- Wuhan National Laboratory for Optoelectronics, Optics Valley Laboratory, School of Optical and Electronic Information, Huazhong University of Science and Technology, Wuhan, China, 430074
| | - Tao Zhang
- Wuhan National Laboratory for Optoelectronics, Optics Valley Laboratory, School of Optical and Electronic Information, Huazhong University of Science and Technology, Wuhan, China, 430074
| | - K. Kirk Shung
- NIH Resource Center for Medical Ultrasonic Transducer Technology, Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Benpeng Zhu
- Wuhan National Laboratory for Optoelectronics, Optics Valley Laboratory, School of Optical and Electronic Information, Huazhong University of Science and Technology, Wuhan, China, 430074
- State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai 200050, China
| |
Collapse
|
12
|
Boudreault F, Tan JJ, Grygorczyk R. Propidium uptake and ATP release in A549 cells share similar transport mechanisms. Biophys J 2022; 121:1593-1609. [PMID: 35398020 PMCID: PMC9117937 DOI: 10.1016/j.bpj.2022.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 03/03/2022] [Accepted: 04/04/2022] [Indexed: 11/02/2022] Open
Abstract
The lipid bilayer of eukaryotic cells' plasma membrane is almost impermeable to small ions and large polar molecules, but its miniscule basal permeability in intact cells is poorly characterized. This report describes the intrinsic membrane permeability of A549 cells toward the charged molecules propidium (Pr2+) and ATP4-. Under isotonic conditions, we detected with quantitative fluorescence microscopy, a continuous low-rate uptake of Pr (∼150 × 10-21 moles (zmol)/h/cell, [Pr]o = 150 μM, 32°C). It was stimulated transiently but strongly by 66% hypotonic cell swelling reaching an influx amplitude of ∼1500 (zmol/h)/cell. The progressive Pr uptake with increasing [Pr]o (30, 150, and 750 μM) suggested a permeation mechanism by simple diffusion. We quantified separately ATP release with custom wide-field-of-view chemiluminescence imaging. The strong proportionality between ATP efflux and Pr2+ influx during hypotonic challenge, and the absence of stimulation of transmembrane transport following 300% hypertonic shock, indicated that ATP and Pr travel the same conductive pathway. The fluorescence images revealed a homogeneously distributed intracellular uptake of Pr not consistent with high-conductance channels expressed at low density on the plasma membrane. We hypothesized that the pathway consists of transiently formed water pores evenly spread across the plasma membrane. The abolition of cell swelling-induced Pr uptake with 500 μM gadolinium, a known modulator of membrane fluidity, supported the involvement of water pores whose formation depends on the membrane fluidity. Our study suggests an alternative model of a direct permeation of ATP (and other molecules) through the phospholipid bilayer, which may have important physiological implications.
Collapse
|
13
|
Rich J, Tian Z, Huang TJ. Sonoporation: Past, Present, and Future. ADVANCED MATERIALS TECHNOLOGIES 2022; 7:2100885. [PMID: 35399914 PMCID: PMC8992730 DOI: 10.1002/admt.202100885] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Indexed: 05/09/2023]
Abstract
A surge of research in intracellular delivery technologies is underway with the increased innovations in cell-based therapies and cell reprogramming. Particularly, physical cell membrane permeabilization techniques are highlighted as the leading technologies because of their unique features, including versatility, independence of cargo properties, and high-throughput delivery that is critical for providing the desired cell quantity for cell-based therapies. Amongst the physical permeabilization methods, sonoporation holds great promise and has been demonstrated for delivering a variety of functional cargos, such as biomolecular drugs, proteins, and plasmids, to various cells including cancer, immune, and stem cells. However, traditional bubble-based sonoporation methods usually require special contrast agents. Bubble-based sonoporation methods also have high chances of inducing irreversible damage to critical cell components, lowering the cell viability, and reducing the effectiveness of delivered cargos. To overcome these limitations, several novel non-bubble-based sonoporation mechanisms are under development. This review will cover both the bubble-based and non-bubble-based sonoporation mechanisms being employed for intracellular delivery, the technologies being investigated to overcome the limitations of traditional platforms, as well as perspectives on the future sonoporation mechanisms, technologies, and applications.
Collapse
Affiliation(s)
- Joseph Rich
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Zhenhua Tian
- Department of Aerospace Engineering, Mississippi State University, Mississippi State, MS, 39762, USA
| | - Tony Jun Huang
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, 27708, USA
| |
Collapse
|
14
|
Yoo J, Kim H, Kim Y, Lim HG, Kim HH. Collapse pressure measurement of single hollow glass microsphere using single-beam acoustic tweezer. ULTRASONICS SONOCHEMISTRY 2022; 82:105844. [PMID: 34965507 PMCID: PMC8799605 DOI: 10.1016/j.ultsonch.2021.105844] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 11/08/2021] [Accepted: 11/21/2021] [Indexed: 06/14/2023]
Abstract
Microbubbles are widely used in medical ultrasound imaging and drug delivery. Many studies have attempted to quantify the collapse pressure of microbubbles using methods that vary depending on the type and population of bubbles and the frequency band of the ultrasound. However, accurate measurement of collapse pressure is difficult as a result of non-acoustic pressure factors generated by physical and chemical reactions such as dissolution, cavitation, and interaction between bubbles. In this study, we developed a method for accurately measuring collapse pressure using only ultrasound pulse acoustic pressure. Under the proposed method, the collapse pressure of a single hollow glass microsphere (HGM) is measured using a high-frequency (20-40 MHz) single-beam acoustic tweezer (SBAT), thereby eliminating the influence of additional factors. Based on these measurements, the collapse pressure is derived as a function of the HGM size using the microspheres' true density. We also developed a method for estimating high-frequency acoustic pressure, whose measurement using current hydrophone equipment is complicated by limitations in the size of the active aperture. By recording the transmit voltage at the moment of collapse and referencing it against the corresponding pressure, it is possible to estimate the acoustic pressure at the given transmit condition. These results of this study suggest a method for quantifying high-frequency acoustic pressure, provide a potential reference for the characterization of bubble collapse pressure, and demonstrate the potential use of acoustic tweezers as a tool for measuring the elastic properties of particles/cells.
Collapse
Affiliation(s)
- Jinhee Yoo
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang 37673, Republic of Korea; Medical Device Innovation Center, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Hyunhee Kim
- Medical Device Innovation Center, Pohang University of Science and Technology, Pohang 37673, Republic of Korea; Department of Convergence IT Engineering, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Yeonggeun Kim
- Medical Device Innovation Center, Pohang University of Science and Technology, Pohang 37673, Republic of Korea; Department of Convergence IT Engineering, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Hae Gyun Lim
- Department of Biomedical Engineering, Pukyong National University, Busan 48513, Republic of Korea.
| | - Hyung Ham Kim
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang 37673, Republic of Korea; Medical Device Innovation Center, Pohang University of Science and Technology, Pohang 37673, Republic of Korea; Department of Convergence IT Engineering, Pohang University of Science and Technology, Pohang 37673, Republic of Korea; Department of Electrical Engineering, Pohang University of Science and Technology, Pohang 37673, Republic of Korea.
| |
Collapse
|
15
|
Chen H, Zhu N, Osman M, Biskowitz R, Liu J, Khandare S, Butler P, Wong PK, Kothapalli SR. A transparent low intensity pulsed ultrasound (LIPUS) chip for high-throughput cell stimulation. LAB ON A CHIP 2021; 21:4734-4742. [PMID: 34739019 DOI: 10.1039/d1lc00667c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
We report an on-chip platform for low-intensity pulsed ultrasound (LIPUS) stimulation of cells directly cultured on a biocompatible surface of a transparent ultrasound transducer (TUT) fabricated using lithium niobate. The high light transmittance (>80%) and compact size (3 mm × 3 mm × 2 mm) of TUTs allowed easy integration with powerful optical microscopy techniques with no additional acoustic coupling and risk for contamination. TUTs were excited with varying acoustic excitation parameters (voltage amplitude and duty cycle) and resulting live cell calcium signaling was simultaneously imaged using time-lapse confocal microscopy, while the temperature change was measured by a thermocouple. Quantitative single-cell fluorescence analysis revealed the dynamic calcium signaling responses and together with the temperature measurements elucidated the optimal stimulation parameters for non-thermal and thermal effects. The fluorescence change profile was distinct from the recorded temperature change (<1 degree Celsius) profile under LIPUS treatment conditions. Cell dead assay results confirmed cells remain viable after the LIPUS treatment. These results confirmed that the TUT platform enables controllable, safe, high-throughput, and uniform mechanical stimulation of all plated cells. The on-chip LIPUS stimulation using TUTs has the potential to attract several in vitro and in vivo biomedical applications such as controlling stem cell differentiation and proliferation, studying biomechanical properties of cancer cells, and gaining fundamental insights into mechanotransduction pathways when integrated with state-of-the-art high-speed and high-resolution microscopy techniques.
Collapse
Affiliation(s)
- Haoyang Chen
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA.
| | - Ninghao Zhu
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA.
| | - Mohamed Osman
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA.
| | - Ryan Biskowitz
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA.
| | - Jinyun Liu
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA.
| | - Shubham Khandare
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA.
| | - Peter Butler
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA.
| | - Pak Kin Wong
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA.
- Department of Mechanical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Sri-Rajasekhar Kothapalli
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA.
- Penn State Cancer Institute, The Pennsylvania State University, Hershey, PA 17033, USA
| |
Collapse
|
16
|
Advances in engineering and synthetic biology toward improved therapeutic immune cells. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2021. [DOI: 10.1016/j.cobme.2021.100342] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
17
|
Zhang N, Wang J, Foiret J, Dai Z, Ferrara KW. Synergies between therapeutic ultrasound, gene therapy and immunotherapy in cancer treatment. Adv Drug Deliv Rev 2021; 178:113906. [PMID: 34333075 PMCID: PMC8556319 DOI: 10.1016/j.addr.2021.113906] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/12/2021] [Accepted: 07/25/2021] [Indexed: 12/14/2022]
Abstract
Due to the ease of use and excellent safety profile, ultrasound is a promising technique for both diagnosis and site-specific therapy. Ultrasound-based techniques have been developed to enhance the pharmacokinetics and efficacy of therapeutic agents in cancer treatment. In particular, transfection with exogenous nucleic acids has the potential to stimulate an immune response in the tumor microenvironment. Ultrasound-mediated gene transfection is a growing field, and recent work has incorporated this technique into cancer immunotherapy. Compared with other gene transfection methods, ultrasound-mediated gene transfection has a unique opportunity to augment the intracellular uptake of nucleic acids while safely and stably modulating the expression of immunostimulatory cytokines. The development and commercialization of therapeutic ultrasound systems further enhance the potential translation. In this Review, we introduce the underlying mechanisms and ongoing preclinical studies of ultrasound-based techniques in gene transfection for cancer immunotherapy. Furthermore, we expand on aspects of therapeutic ultrasound that impact gene therapy and immunotherapy, including tumor debulking, enhancing cytokines and chemokines and altering nanoparticle pharmacokinetics as these effects of ultrasound cannot be fully dissected from targeted gene therapy. We finally explore the outlook for this rapidly developing field.
Collapse
Affiliation(s)
- Nisi Zhang
- Department of Radiology, Stanford University, Palo Alto, CA, USA; Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China
| | - James Wang
- Department of Radiology, Stanford University, Palo Alto, CA, USA
| | - Josquin Foiret
- Department of Radiology, Stanford University, Palo Alto, CA, USA
| | - Zhifei Dai
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China.
| | | |
Collapse
|
18
|
Hao R, Yu Z, Du J, Hu S, Yuan C, Guo H, Zhang Y, Yang H. A High-Throughput Nanofluidic Device for Exosome Nanoporation to Develop Cargo Delivery Vehicles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2102150. [PMID: 34291570 DOI: 10.1002/smll.202102150] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/15/2021] [Indexed: 06/13/2023]
Abstract
Efficient loading of various exogenous cargos into exosomes while not affecting their integrity and functionalities remains a major challenge. Here, a nanofluidic device named "exosome nanoporator (ENP)" is presented for high-throughput loading of various cargos into exosomes. By transporting exosomes through nanochannels with height comparable to their dimension, exosome membranes are permeabilized by mechanical compression and fluid shear, allowing the influx of cargo molecules into the exosomes from the surrounding solution while maintaining exosome integrity. The ENP consisting of an array of 30 000 nanochannels demonstrates a high sample throughput, and the working mechanism of the device is elucidated through experimental and numerical study. Further, the exosomes treated by the ENP can deliver their drug cargos to human non-small cell lung cancer cells and induce cell death, indicating the potential opportunities of the device for developing new exosome-based delivery vehicles for medical and biological applications.
Collapse
Affiliation(s)
- Rui Hao
- Laboratory of Biomedical Microsystems and Nano Devices, Center for Bionic Sensing and Intelligence, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen, 361005, China
| | - Zitong Yu
- Laboratory of Biomedical Microsystems and Nano Devices, Center for Bionic Sensing and Intelligence, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Jing Du
- Laboratory of Biomedical Microsystems and Nano Devices, Center for Bionic Sensing and Intelligence, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Shi Hu
- Laboratory of Biomedical Microsystems and Nano Devices, Center for Bionic Sensing and Intelligence, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Chao Yuan
- Laboratory of Biomedical Microsystems and Nano Devices, Center for Bionic Sensing and Intelligence, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Hang Guo
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen, 361005, China
| | - Yi Zhang
- Center for Medical AI, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Hui Yang
- Laboratory of Biomedical Microsystems and Nano Devices, Center for Bionic Sensing and Intelligence, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| |
Collapse
|
19
|
Hur J, Chung AJ. Microfluidic and Nanofluidic Intracellular Delivery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2004595. [PMID: 34096197 PMCID: PMC8336510 DOI: 10.1002/advs.202004595] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 04/14/2021] [Indexed: 05/05/2023]
Abstract
Innate cell function can be artificially engineered and reprogrammed by introducing biomolecules, such as DNAs, RNAs, plasmid DNAs, proteins, or nanomaterials, into the cytosol or nucleus. This process of delivering exogenous cargos into living cells is referred to as intracellular delivery. For instance, clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 gene editing begins with internalizing Cas9 protein and guide RNA into cells, and chimeric antigen receptor-T (CAR-T) cells are prepared by delivering CAR genes into T lymphocytes for cancer immunotherapies. To deliver external biomolecules into cells, tools, including viral vectors, and electroporation have been traditionally used; however, they are suboptimal for achieving high levels of intracellular delivery while preserving cell viability, phenotype, and function. Notably, as emerging solutions, microfluidic and nanofluidic approaches have shown remarkable potential for addressing this open challenge. This review provides an overview of recent advances in microfluidic and nanofluidic intracellular delivery strategies and discusses new opportunities and challenges for clinical applications. Furthermore, key considerations for future efforts to develop microfluidics- and nanofluidics-enabled next-generation intracellular delivery platforms are outlined.
Collapse
Affiliation(s)
- Jeongsoo Hur
- School of Biomedical EngineeringKorea UniversitySeoul02841Republic of Korea
| | - Aram J. Chung
- School of Biomedical EngineeringInterdisciplinary Program in Precision Public HealthKorea UniversitySeoul02841Republic of Korea
| |
Collapse
|
20
|
Kim S, Moon S, Rho S, Yoon S. Measurements of acoustic radiation force of ultrahigh frequency ultrasonic transducers using model-based approach. APPLIED PHYSICS LETTERS 2021; 118:184102. [PMID: 33981116 PMCID: PMC8096457 DOI: 10.1063/5.0044512] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 04/19/2021] [Indexed: 05/13/2023]
Abstract
Even though ultrahigh frequency ultrasonic transducers over 60 MHz have been used for single-cell-level manipulation such as intracellular delivery, acoustic tweezers, and stimulation to investigate cell phenotype and cell mechanics, no techniques have been available to measure the actual acoustic radiation force (ARF) applied to target cells. Therefore, we have developed an approach to measure the ARF of ultrahigh frequency ultrasonic transducers using a theoretical model of the dynamics of a solid sphere in a gelatin phantom. To estimate ARF at the focus of a 130 MHz transducer, we matched measured maximum displacements of a solid sphere with theoretical calculations. We selected appropriate ranges of input voltages and pulse durations for single-cell applications, and the estimated ARF was in the range of tens of μN. To gauge the influence of pulse duration, an impulse of different pulse durations was estimated. Fluorescence resonance energy transfer live cell imaging was demonstrated to visualize calcium transport between cells after a target single cell was stimulated by the developed ultrasonic transducer.
Collapse
Affiliation(s)
| | | | | | - Sangpil Yoon
- Author to whom correspondence should be addressed: . Tel.: +1-514-631‐6510
| |
Collapse
|
21
|
Ramesan S, Rezk AR, Cevaal PM, Cortez-Jugo C, Symons J, Yeo LY. Acoustofection: High-Frequency Vibrational Membrane Permeabilization for Intracellular siRNA Delivery into Nonadherent Cells. ACS APPLIED BIO MATERIALS 2021; 4:2781-2789. [PMID: 35014317 DOI: 10.1021/acsabm.1c00003] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The internalization of therapeutic molecules into cells-a critical step in enabling a suite of autologous ex vivo gene and cell therapies-is highly regulated by the lipid barrier imposed by the cell membrane. Strategies to increase the efficiency of delivering these exogenous payloads into the cell, while maintaining the integrity of both the therapeutic molecules to be delivered as well as the host cells they are delivered to, are therefore required. This is especially the case for suspension cells that are particularly difficult to transfect. In this work, we show that it is possible to enhance the uptake of short interfering RNA (siRNA) into nonadherent Jurkat and HuT 78 cells with a rapid poration-free method involving high-frequency (MHz order) acoustic excitation. The 2-fold enhancement in gene knockdown is almost comparable with that obtained with conventional nucleofection, which is among the most widely used intracellular delivery methods, but with considerably higher cell viabilities (>91% compared to approximately 76%) owing to the absence of pore formation. The rapid and effective delivery afforded by the platform, together with its low cost and scalability, therefore renders it a potent tool in the cell engineering pipeline.
Collapse
Affiliation(s)
- Shwathy Ramesan
- Micro/Nanophysics Research Laboratory, School of Engineering, RMIT University, Melbourne, VIC 3000, Australia
| | - Amgad R Rezk
- Micro/Nanophysics Research Laboratory, School of Engineering, RMIT University, Melbourne, VIC 3000, Australia
| | - Paula M Cevaal
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne and Royal Melbourne Hospital, Melbourne, VIC 3000, Australia
| | - Christina Cortez-Jugo
- Department of Chemical Engineering, University of Melbourne, Parkville, VIC 3010, Australia
| | - Jori Symons
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne and Royal Melbourne Hospital, Melbourne, VIC 3000, Australia
| | - Leslie Y Yeo
- Micro/Nanophysics Research Laboratory, School of Engineering, RMIT University, Melbourne, VIC 3000, Australia
| |
Collapse
|
22
|
Yoon S, Pan Y, Shung K, Wang Y. FRET-Based Ca 2+ Biosensor Single Cell Imaging Interrogated by High-Frequency Ultrasound. SENSORS 2020; 20:s20174998. [PMID: 32899249 PMCID: PMC7506572 DOI: 10.3390/s20174998] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/26/2020] [Accepted: 08/31/2020] [Indexed: 11/22/2022]
Abstract
Fluorescence resonance energy transfer (FRET)-based biosensors have advanced live cell imaging by dynamically visualizing molecular events with high temporal resolution. FRET-based biosensors with spectrally distinct fluorophore pairs provide clear contrast between cells during dual FRET live cell imaging. Here, we have developed a new FRET-based Ca2+ biosensor using EGFP and FusionRed fluorophores (FRET-GFPRed). Using different filter settings, the developed biosensor can be differentiated from a typical FRET-based Ca2+ biosensor with ECFP and YPet (YC3.6 FRET Ca2+ biosensor, FRET-CFPYPet). A high-frequency ultrasound (HFU) with a carrier frequency of 150 MHz can target a subcellular region due to its tight focus smaller than 10 µm. Therefore, HFU offers a new single cell stimulations approach for FRET live cell imaging with precise spatial resolution and repeated stimulation for longitudinal studies. Furthermore, the single cell level intracellular delivery of a desired FRET-based biosensor into target cells using HFU enables us to perform dual FRET imaging of a cell pair. We show that a cell pair is defined by sequential intracellular delivery of the developed FRET-GFPRed and FRET-CFPYPet into two target cells using HFU. We demonstrate that a FRET-GFPRed exhibits consistent 10–15% FRET response under typical ionomycin stimulation as well as under a new stimulation strategy with HFU.
Collapse
Affiliation(s)
- Sangpil Yoon
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
- Correspondence: ; Tel.: +1-514-631-6510
| | - Yijia Pan
- Department of Bioengineering, University of California, San Diego, CA 92092, USA; (Y.P.); (Y.W.)
| | - Kirk Shung
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA;
| | - Yingxiao Wang
- Department of Bioengineering, University of California, San Diego, CA 92092, USA; (Y.P.); (Y.W.)
| |
Collapse
|
23
|
Therapeutic Potentials of Localized Blood-Brain Barrier Disruption by Noninvasive Transcranial Focused Ultrasound: A Technical Review. J Clin Neurophysiol 2020; 37:104-117. [PMID: 32142021 DOI: 10.1097/wnp.0000000000000488] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
The demands for region-specific, noninvasive therapies for neurologic/psychiatric conditions are growing. The rise of transcranial focused ultrasound technology has witnessed temporary and reversible disruptions of the blood-brain barrier in the brain with exceptional control over the spatial precisions and depth, all in a noninvasive manner. Starting with small animal studies about a decade ago, the technique is now being explored in nonhuman primates and humans for the assessment of its efficacy and safety. The ability to transfer exogenous/endogenous therapeutic agents, cells, and biomolecules across the blood-brain barrier opens up new therapeutic avenues for various neurologic conditions, with a possibility to modulate the excitability of regional brain function. This review addresses the technical fundamentals, sonication parameters, experimental protocols, and monitoring techniques to examine the efficacy/safety in focused ultrasound-mediated blood-brain barrier disruption and discuss its potential translations to clinical use.
Collapse
|
24
|
Rezk AR, Ahmed H, Ramesan S, Yeo LY. High Frequency Sonoprocessing: A New Field of Cavitation-Free Acoustic Materials Synthesis, Processing, and Manipulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 8:2001983. [PMID: 33437572 PMCID: PMC7788597 DOI: 10.1002/advs.202001983] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 09/17/2020] [Indexed: 04/14/2023]
Abstract
Ultrasound constitutes a powerful means for materials processing. Similarly, a new field has emerged demonstrating the possibility for harnessing sound energy sources at considerably higher frequencies (10 MHz to 1 GHz) compared to conventional ultrasound (⩽3 MHz) for synthesizing and manipulating a variety of bulk, nanoscale, and biological materials. At these frequencies and the typical acoustic intensities employed, cavitation-which underpins most sonochemical or, more broadly, ultrasound-mediated processes-is largely absent, suggesting that altogether fundamentally different mechanisms are at play. Examples include the crystallization of novel morphologies or highly oriented structures; exfoliation of 2D quantum dots and nanosheets; polymer nanoparticle synthesis and encapsulation; and the possibility for manipulating the bandgap of 2D semiconducting materials or the lipid structure that makes up the cell membrane, the latter resulting in the ability to enhance intracellular molecular uptake. These fascinating examples reveal how the highly nonlinear electromechanical coupling associated with such high-frequency surface vibration gives rise to a variety of static and dynamic charge generation and transfer effects, in addition to molecular ordering, polarization, and assembly-remarkably, given the vast dimensional separation between the acoustic wavelength and characteristic molecular length scales, or between the MHz-order excitation frequencies and typical THz-order molecular vibration frequencies.
Collapse
Affiliation(s)
- Amgad R. Rezk
- Micro/Nanophysics Research LaboratorySchool of EngineeringRMIT UniversityMelbourneVIC3000Australia
| | - Heba Ahmed
- Micro/Nanophysics Research LaboratorySchool of EngineeringRMIT UniversityMelbourneVIC3000Australia
| | - Shwathy Ramesan
- Micro/Nanophysics Research LaboratorySchool of EngineeringRMIT UniversityMelbourneVIC3000Australia
| | - Leslie Y. Yeo
- Micro/Nanophysics Research LaboratorySchool of EngineeringRMIT UniversityMelbourneVIC3000Australia
| |
Collapse
|
25
|
Abstract
From basic studies in understanding the role of signaling pathways to therapeutic applications in engineering new cellular functions, efficient and safe techniques to monitor and modulate molecular targets from cells to organs have been extensively developed. The developmental advancement of engineering devices such as microscope and ultrasonic transducers allows us to investigate biological processes at different scales. Synthetic biology has further emerged recently as a powerful platform for the development of new diagnostic and therapeutic molecular tools. The synergetic amalgamation between engineering tools and synthetic biology has rapidly become a new front in the field of bioengineering and biotechnology. In this review, ultrasound and its generated mechanical perturbation are introduced to serve as a non-invasive engineering approach and, integrated with synthetic biology, to remotely control signaling and genetic activities for the guidance of cellular functions deep inside tissue with high spatiotemporal resolutions. This ultrasound-based approach together with synthetic biology has been applied in immunotherapy, neuroscience, and gene delivery, paving the way for the development of next-generation therapeutic tools.
Collapse
Affiliation(s)
- Yijia Pan
- Department of Bioengineering, University of California, San Diego
| | - Sangpil Yoon
- Department of Aerospace and Mechanical Engineering, University of Notre Dame
- Department of Biomedical Engineering, University of Southern California
| | - Linshan Zhu
- Department of Bioengineering, University of California, San Diego
| | - Yingxiao Wang
- Department of Bioengineering, University of California, San Diego
| |
Collapse
|
26
|
Capozza R, Caprettini V, Gonano CA, Bosca A, Moia F, Santoro F, De Angelis F. Cell Membrane Disruption by Vertical Micro-/Nanopillars: Role of Membrane Bending and Traction Forces. ACS APPLIED MATERIALS & INTERFACES 2018; 10:29107-29114. [PMID: 30081625 PMCID: PMC6117743 DOI: 10.1021/acsami.8b08218] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Gaining access to the cell interior is fundamental for many applications, such as electrical recording and drug and biomolecular delivery. A very promising technique consists of culturing cells on micro-/nanopillars. The tight adhesion and high local deformation of cells in contact with nanostructures can promote the permeabilization of lipids at the plasma membrane, providing access to the internal compartment. However, there is still much experimental controversy regarding when and how the intracellular environment is targeted and the role of the geometry and interactions with surfaces. Consequently, we investigated, by coarse-grained molecular dynamics simulations of the cell membrane, the mechanical properties of the lipid bilayer under high strain and bending conditions. We found out that a high curvature of the lipid bilayer dramatically lowers the traction force necessary to achieve membrane rupture. Afterward, we experimentally studied the permeabilization rate of the cell membrane by pillars with comparable aspect ratios but different sharpness values at the edges. The experimental data support the simulation results: even pillars with diameters in the micron range may cause local membrane disruption when their edges are sufficiently sharp. Therefore, the permeabilization likelihood is connected to the local geometric features of the pillars rather than diameter or aspect ratio. The present study can also provide significant contributions to the design of three-dimensional biointerfaces for tissue engineering and cellular growth.
Collapse
Affiliation(s)
- Rosario Capozza
- Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Valeria Caprettini
- Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
- Università
degli studi di Genova, Genova 16126, Italy
| | - Carlo A. Gonano
- Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Alessandro Bosca
- Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Fabio Moia
- Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Francesca Santoro
- Center
for Advanced Biomaterials for Healthcare, Istituto Italiano di Tecnologia, 80125 Napoli, Italy
| | | |
Collapse
|
27
|
Stewart MP, Langer R, Jensen KF. Intracellular Delivery by Membrane Disruption: Mechanisms, Strategies, and Concepts. Chem Rev 2018; 118:7409-7531. [PMID: 30052023 PMCID: PMC6763210 DOI: 10.1021/acs.chemrev.7b00678] [Citation(s) in RCA: 456] [Impact Index Per Article: 65.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Intracellular delivery is a key step in biological research and has enabled decades of biomedical discoveries. It is also becoming increasingly important in industrial and medical applications ranging from biomanufacture to cell-based therapies. Here, we review techniques for membrane disruption-based intracellular delivery from 1911 until the present. These methods achieve rapid, direct, and universal delivery of almost any cargo molecule or material that can be dispersed in solution. We start by covering the motivations for intracellular delivery and the challenges associated with the different cargo types-small molecules, proteins/peptides, nucleic acids, synthetic nanomaterials, and large cargo. The review then presents a broad comparison of delivery strategies followed by an analysis of membrane disruption mechanisms and the biology of the cell response. We cover mechanical, electrical, thermal, optical, and chemical strategies of membrane disruption with a particular emphasis on their applications and challenges to implementation. Throughout, we highlight specific mechanisms of membrane disruption and suggest areas in need of further experimentation. We hope the concepts discussed in our review inspire scientists and engineers with further ideas to improve intracellular delivery.
Collapse
Affiliation(s)
- Martin P. Stewart
- Department of Chemical Engineering, Massachusetts Institute
of Technology, Cambridge, USA
- The Koch Institute for Integrative Cancer Research,
Massachusetts Institute of Technology, Cambridge, USA
| | - Robert Langer
- Department of Chemical Engineering, Massachusetts Institute
of Technology, Cambridge, USA
- The Koch Institute for Integrative Cancer Research,
Massachusetts Institute of Technology, Cambridge, USA
| | - Klavs F. Jensen
- Department of Chemical Engineering, Massachusetts Institute
of Technology, Cambridge, USA
| |
Collapse
|
28
|
Kim MG, Park J, Zhou Q, Shung KK. Novel Configurations of Ultrahigh Frequency (≤600 MHz) Analog Frontend for High Resolution Ultrasound Measurement. SENSORS (BASEL, SWITZERLAND) 2018; 18:s18082598. [PMID: 30096810 PMCID: PMC6111284 DOI: 10.3390/s18082598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 08/04/2018] [Accepted: 08/07/2018] [Indexed: 05/16/2023]
Abstract
In this article, an approach to designing and developing an ultrahigh frequency (≤600 MHz) ultrasound analog frontend with Golay coded excitation sequence for high resolution imaging applications is presented. For the purpose of visualizing specific structures or measuring functional responses of micron-sized biological samples, a higher frequency ultrasound is needed to obtain a decent spatial resolution while it lowers the signal-to-noise ratio, the difference in decibels between the signal level and the background noise level, due to the higher attenuation coefficient. In order to enhance the signal-to-noise ratio, conventional approach was to increase the transmit voltage level. However, it may cause damaging the extremely thin piezoelectric material in the ultrahigh frequency range. In this paper, we present a novel design of ultrahigh frequency (≤600 MHz) frontend system capable of performing pseudo Golay coded excitation by configuring four independently operating pulse generators in parallel and the consecutive delayed transmission from each channel. Compared with the conventional monocycle pulse approach, the signal-to-noise ratio of the proposed approach was improved by 7⁻9 dB without compromising the spatial resolution. The measured axial and lateral resolutions of wire targets were 16.4 µm and 10.6 µm by using 156 MHz 4 bit pseudo Golay coded excitation, respectively and 4.5 µm and 7.7 µm by using 312 MHz 4 bit pseudo Golay coded excitation, respectively.
Collapse
Affiliation(s)
- Min Gon Kim
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA.
| | - Jinhyoung Park
- Department of Biomedical Engineering, Sungkyunkwan University, Suwon, Gyeonggi-do 16419, Korea.
| | - Qifa Zhou
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA.
| | - Koping Kirk Shung
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
29
|
Yu J, Chen Z, Yan F. Advances in mechanism studies on ultrasonic gene delivery at cellular level. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2018; 142:1-9. [PMID: 30031881 DOI: 10.1016/j.pbiomolbio.2018.07.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 07/15/2018] [Accepted: 07/19/2018] [Indexed: 01/23/2023]
Abstract
Ultrasound provides a means for intracellular gene delivery, contributing to a noninvasive and spatiotemporally controllable strategy suitable for clinical applications. Many studies have been done to provide mechanisms of ultrasound-mediated gene delivery at the cellular level. This review summarizes the studies on the important aspects of the mechanisms, providing an overview of recent progress in cellular experiment of ultrasound-mediated gene delivery.
Collapse
Affiliation(s)
- Jinsui Yu
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, PR China
| | - Zhiyi Chen
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, PR China.
| | - Fei Yan
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China.
| |
Collapse
|
30
|
Ramesan S, Rezk AR, Dekiwadia C, Cortez-Jugo C, Yeo LY. Acoustically-mediated intracellular delivery. NANOSCALE 2018; 10:13165-13178. [PMID: 29964280 DOI: 10.1039/c8nr02898b] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Recent breakthroughs in gene editing have necessitated practical ex vivo methods to rapidly and efficiently re-engineer patient-harvested cells. Many physical membrane-disruption or pore-forming techniques for intracellular delivery, however, result in poor cell viability, while most carrier-mediated techniques suffer from suboptimal endosomal escape and hence cytoplasmic or nuclear targeting. In this work, we show that short exposure of cells to high frequency (>10 MHz) acoustic excitation facilitates temporal reorganisation of the lipid structure in the cell membrane that enhances translocation of gold nanoparticles and therapeutic molecules into the cell within just ten minutes. Due to its transient nature, rapid cell self-healing is observed, leading to high cellular viabilities (>97%). Moreover, the internalised cargo appears to be uniformly distributed throughout the cytosol, circumventing the need for strategies to facilitate endosomal escape. In the case of siRNA delivery, the method is seen to enhance gene silencing by over twofold, demonstrating its potential for enhancing therapeutic delivery into cells.
Collapse
Affiliation(s)
- Shwathy Ramesan
- Micro/Nanophysics Research Laboratory, School of Engineering, RMIT University, Melbourne, VIC 3000, Australia.
| | - Amgad R Rezk
- Micro/Nanophysics Research Laboratory, School of Engineering, RMIT University, Melbourne, VIC 3000, Australia.
| | - Chaitali Dekiwadia
- RMIT Microscopy and Microanalysis Facility, RMIT University, Melbourne, VIC 3000, Australia
| | - Christina Cortez-Jugo
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology & Department of Chemical and Biomolecular Engineering, University of Melbourne, Parkville, VIC 3010, Australia
| | - Leslie Y Yeo
- Micro/Nanophysics Research Laboratory, School of Engineering, RMIT University, Melbourne, VIC 3000, Australia.
| |
Collapse
|
31
|
Kim MG, Yoon S, Chiu CT, Shung KK. Investigation of Optimized Treatment Conditions for Acoustic-Transfection Technique for Intracellular Delivery of Macromolecules. ULTRASOUND IN MEDICINE & BIOLOGY 2018; 44:622-634. [PMID: 29284555 PMCID: PMC5800999 DOI: 10.1016/j.ultrasmedbio.2017.11.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 11/09/2017] [Accepted: 11/13/2017] [Indexed: 05/03/2023]
Abstract
Manipulation of cellular functions and structures by introduction of genetic materials inside cells has been one of the most prominent research areas in biomedicine. High-frequency ultrasound acoustic-transfection has recently been developed and confirmed by intracellular delivery of small molecules into HeLa cells at the single-cell level with high cell viability. After we proved the concept underlying the acoustic-transfection technique, treatment conditions for different human cancer cell lines have been intensively investigated to further develop acoustic-transfection as a versatile and adaptable transfection method by satisfying the requirements of high-delivery efficiency and cell membrane permeability with minimal membrane disruption. To determine optimal treatment conditions for different cell lines, we developed a quantitative intracellular delivery score based on delivery efficiency, cell membrane permeability and cell viability after 4 and 20 h of treatment. The intracellular delivery of macromolecules and the simultaneous intracellular delivery of two molecules under optimal treatment conditions were successfully achieved. We found that DNA plasmid was delivered by acoustic-transfection technique into epiblast stem cells, which expressed transient mCherry fluorescence.
Collapse
Affiliation(s)
- Min Gon Kim
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| | - Sangpil Yoon
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA.
| | - Chi Tat Chiu
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| | - K Kirk Shung
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
32
|
Meacham JM, Durvasula K, Degertekin FL, Fedorov AG. Enhanced intracellular delivery via coordinated acoustically driven shear mechanoporation and electrophoretic insertion. Sci Rep 2018; 8:3727. [PMID: 29487375 PMCID: PMC5829135 DOI: 10.1038/s41598-018-22042-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 02/15/2018] [Indexed: 12/13/2022] Open
Abstract
Delivery of large and structurally complex target molecules into cells is vital to the emerging areas of cellular modification and molecular therapy. Inadequacy of prevailing in vivo (viral) and in vitro (liposomal) gene transfer methods for delivery of proteins and a growing diversity of synthetic nanomaterials has encouraged development of alternative physical approaches. Efficacy of injury/diffusion-based delivery via shear mechanoporation is largely insensitive to cell type and target molecule; however, enhanced flexibility is typically accompanied by reduced gene transfer effectiveness. We detail a method to improve transfection efficiency through coordinated mechanical disruption of the cell membrane and electrophoretic insertion of DNA to the cell interior. An array of micromachined nozzles focuses ultrasonic pressure waves, creating a high-shear environment that promotes transient pore formation in membranes of transmitted cells. Acoustic Shear Poration (ASP) allows passive cytoplasmic delivery of small to large nongene macromolecules into established and primary cells at greater than 75% efficiency. Addition of an electrophoretic action enables active transport of target DNA molecules to substantially augment transfection efficiency of passive mechanoporation/diffusive delivery without affecting viability. This two-stage poration/insertion method preserves the compelling flexibility of shear-based delivery, yet substantially enhances capabilities for active transport and transfection of plasmid DNA.
Collapse
Affiliation(s)
- J Mark Meacham
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, MO, 63130, USA.
| | | | - F Levent Degertekin
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA.,Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA.,G. W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Andrei G Fedorov
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA.,G. W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| |
Collapse
|
33
|
Mechanogenetics for the remote and noninvasive control of cancer immunotherapy. Proc Natl Acad Sci U S A 2018; 115:992-997. [PMID: 29343642 DOI: 10.1073/pnas.1714900115] [Citation(s) in RCA: 176] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
While cell-based immunotherapy, especially chimeric antigen receptor (CAR)-expressing T cells, is becoming a paradigm-shifting therapeutic approach for cancer treatment, there is a lack of general methods to remotely and noninvasively regulate genetics in live mammalian cells and animals for cancer immunotherapy within confined local tissue space. To address this limitation, we have identified a mechanically sensitive Piezo1 ion channel (mechanosensor) that is activatable by ultrasound stimulation and integrated it with engineered genetic circuits (genetic transducer) in live HEK293T cells to convert the ultrasound-activated Piezo1 into transcriptional activities. We have further engineered the Jurkat T-cell line and primary T cells (peripheral blood mononuclear cells) to remotely sense the ultrasound wave and transduce it into transcriptional activation for the CAR expression to recognize and eradicate target tumor cells. This approach is modular and can be extended for remote-controlled activation of different cell types with high spatiotemporal precision for therapeutic applications.
Collapse
|
34
|
Caprettini V, Cerea A, Melle G, Lovato L, Capozza R, Huang JA, Tantussi F, Dipalo M, De Angelis F. Soft electroporation for delivering molecules into tightly adherent mammalian cells through 3D hollow nanoelectrodes. Sci Rep 2017; 7:8524. [PMID: 28819252 PMCID: PMC5561120 DOI: 10.1038/s41598-017-08886-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 07/14/2017] [Indexed: 12/26/2022] Open
Abstract
Electroporation of in-vitro cultured cells is widely used in biological and medical areas to deliver molecules of interest inside cells. Since very high electric fields are required to electroporate the plasma membrane, depending on the geometry of the electrodes the required voltages can be very high and often critical to cell viability. Furthermore, in traditional electroporation configuration based on planar electrodes there is no a priori certain feedback about which cell has been targeted and delivered and the addition of fluorophores may be needed to gain this information. In this study we present a nanofabricated platform able to perform intracellular delivery of membrane-impermeable molecules by opening transient nanopores into the lipid membrane of adherent cells with high spatial precision and with the application of low voltages (1.5–2 V). This result is obtained by exploiting the tight seal that the cells present with 3D fluidic hollow gold-coated nanostructures that act as nanochannels and nanoelectrodes at the same time. The final soft-electroporation platform provides an accessible approach for controlled and selective drug delivery on ordered arrangements of cells.
Collapse
Affiliation(s)
- Valeria Caprettini
- Istituto Italiano di Tecnologia, Genoa, 16163, Italy.,Università degli studi di Genova, Genoa, 16126, Italy
| | - Andrea Cerea
- Istituto Italiano di Tecnologia, Genoa, 16163, Italy.,Università degli studi di Genova, Genoa, 16126, Italy
| | - Giovanni Melle
- Istituto Italiano di Tecnologia, Genoa, 16163, Italy.,Università degli studi di Genova, Genoa, 16126, Italy
| | - Laura Lovato
- Istituto Italiano di Tecnologia, Genoa, 16163, Italy
| | | | - Jian-An Huang
- Istituto Italiano di Tecnologia, Genoa, 16163, Italy
| | | | | | | |
Collapse
|
35
|
Acoustic-transfection for genomic manipulation of single-cells using high frequency ultrasound. Sci Rep 2017; 7:5275. [PMID: 28706248 PMCID: PMC5509725 DOI: 10.1038/s41598-017-05722-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 06/16/2017] [Indexed: 01/21/2023] Open
Abstract
Efficient intracellular delivery of biologically active macromolecules has been a challenging but important process for manipulating live cells for research and therapeutic purposes. There have been limited transfection techniques that can deliver multiple types of active molecules simultaneously into single-cells as well as different types of molecules into physically connected individual neighboring cells separately with high precision and low cytotoxicity. Here, a high frequency ultrasound-based remote intracellular delivery technique capable of delivery of multiple DNA plasmids, messenger RNAs, and recombinant proteins is developed to allow high spatiotemporal visualization and analysis of gene and protein expressions as well as single-cell gene editing using clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein-9 nuclease (Cas9), a method called acoustic-transfection. Acoustic-transfection has advantages over typical sonoporation because acoustic-transfection utilizing ultra-high frequency ultrasound over 150 MHz can directly deliver gene and proteins into cytoplasm without microbubbles, which enables controlled and local intracellular delivery to acoustic-transfection technique. Acoustic-transfection was further demonstrated to deliver CRISPR-Cas9 systems to successfully modify and reprogram the genome of single live cells, providing the evidence of the acoustic-transfection technique for precise genome editing using CRISPR-Cas9.
Collapse
|
36
|
Lee H, Kim H, Han H, Lee M, Lee S, Yoo H, Chang JH, Kim H. Microbubbles used for contrast enhanced ultrasound and theragnosis: a review of principles to applications. Biomed Eng Lett 2017; 7:59-69. [PMID: 30603152 PMCID: PMC6208473 DOI: 10.1007/s13534-017-0016-5] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Revised: 12/26/2016] [Accepted: 01/18/2017] [Indexed: 12/31/2022] Open
Abstract
Ultrasound was developed several decades ago as a useful imaging modality, and it became the second most popular diagnostic tool due to its non-invasiveness, real-time capabilities, and safety. Additionally, ultrasound has been used as a therapeutic tool with several therapeutic agents and in nanomedicine. Ultrasound imaging is often used to diagnose many types of cancers, including breast, stomach, and thyroid cancers. In addition, ultrasound-mediated therapy is used in cases of joint inflammation, rheumatoid arthritis, and osteoarthritis. Microbubbles, when used as ultrasound contrast agents, can act as echo-enhancers and therapeutic agents, and they can play an essential role in ultrasound imaging and ultrasound-mediated therapy. Recently, various types of ultrasound contrast agents made of lipid, polymer, and protein shells have been used. Air, nitrogen, and perfluorocarbon are usually included in the core of the microbubbles to enhance ultrasound imaging, and therapeutic drugs are conjugated and loaded onto the surface or into the core of the microbubbles, depending on the purpose and properties of the substance. Many research groups have utilized ultrasound contrast agents to enhance the imaging signal in blood vessels or tissues and to overcome the blood-brain barrier or blood-retina barrier. These agents are also used to help treat diseases in various regions or systems of the body, such as the cardiovascular system, or as a cancer treatment. In addition, with the introduction of targeted moiety and multiple functional groups, ultrasound contrast agents are expected to have a potential future in ultrasound imaging and therapy. In this paper, we briefly review the principles of ultrasound and introduce the underlying theory, applications, limitations, and future perspectives of ultrasound contrast agents.
Collapse
Affiliation(s)
- Hohyeon Lee
- Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107 Republic of Korea
| | - Haemin Kim
- Department of Biomedical Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107 Republic of Korea
| | - Hyounkoo Han
- Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107 Republic of Korea
| | - Minji Lee
- Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107 Republic of Korea
| | - Sunho Lee
- Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107 Republic of Korea
| | - Hongkeun Yoo
- Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107 Republic of Korea
| | - Jin Ho Chang
- Department of Biomedical Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107 Republic of Korea
- Sogang Institute of Advanced Technology, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107 Republic of Korea
| | - Hyuncheol Kim
- Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107 Republic of Korea
- Department of Biomedical Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107 Republic of Korea
| |
Collapse
|