1
|
Przystupski D, Ussowicz M. Landscape of Cellular Bioeffects Triggered by Ultrasound-Induced Sonoporation. Int J Mol Sci 2022; 23:ijms231911222. [PMID: 36232532 PMCID: PMC9569453 DOI: 10.3390/ijms231911222] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/16/2022] [Accepted: 09/20/2022] [Indexed: 11/18/2022] Open
Abstract
Sonoporation is the process of transient pore formation in the cell membrane triggered by ultrasound (US). Numerous studies have provided us with firm evidence that sonoporation may assist cancer treatment through effective drug and gene delivery. However, there is a massive gap in the body of literature on the issue of understanding the complexity of biophysical and biochemical sonoporation-induced cellular effects. This study provides a detailed explanation of the US-triggered bioeffects, in particular, cell compartments and the internal environment of the cell, as well as the further consequences on cell reproduction and growth. Moreover, a detailed biophysical insight into US-provoked pore formation is presented. This study is expected to review the knowledge of cellular effects initiated by US-induced sonoporation and summarize the attempts at clinical implementation.
Collapse
|
2
|
Centner CS, Moore JT, Baxter ME, Long ZT, Miller JM, Kovatsenko ES, Xie B, Menze MA, Berson RE, Bates PJ, Yaddanapudi K, Kopechek JA. Acoustofluidic-mediated molecular delivery to human T cells with a three-dimensional-printed flow chamber. THE JOURNAL OF THE ACOUSTICAL SOCIETY OF AMERICA 2021; 150:4534. [PMID: 34972278 DOI: 10.1121/10.0009054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 11/09/2021] [Indexed: 06/14/2023]
Abstract
Cell-based therapies have garnered significant interest to treat cancer and other diseases. Acoustofluidic technologies are in development to improve cell therapy manufacturing by facilitating rapid molecular delivery across the plasma membrane via ultrasound and microbubbles (MBs). In this study, a three-dimensional (3D) printed acoustofluidic device was used to deliver a fluorescent molecule, calcein, to human T cells. Intracellular delivery of calcein was assessed after varying parameters such as MB face charge, MB concentration, flow channel geometry, ultrasound pressure, and delivery time point after ultrasound treatment. MBs with a cationic surface charge caused statistically significant increases in calcein delivery during acoustofluidic treatment compared to MBs with a neutral surface charge (p < 0.001). Calcein delivery was significantly higher with a concentric spiral channel geometry compared to a rectilinear channel geometry (p < 0.001). Additionally, calcein delivery was significantly enhanced at increased ultrasound pressures of 5.1 MPa compared to lower ultrasound pressures between 0-3.8 MPa (p < 0.001). These results demonstrate that a 3D-printed acoustofluidic device can significantly enhance intracellular delivery of biomolecules to T cells, which may be a viable approach to advance cell-based therapies.
Collapse
Affiliation(s)
- Connor S Centner
- Department of Bioengineering, University of Louisville, Louisville, Kentucky 40292, USA
| | - John T Moore
- Department of Bioengineering, University of Louisville, Louisville, Kentucky 40292, USA
| | - Mary E Baxter
- Department of Bioengineering, University of Louisville, Louisville, Kentucky 40292, USA
| | - Zachary T Long
- Department of Bioengineering, University of Louisville, Louisville, Kentucky 40292, USA
| | - Jacob M Miller
- Department of Chemical Engineering, University of Louisville, Louisville, Kentucky 40292, USA
| | | | - Benjamin Xie
- Department of Biology, University of Louisville, Louisville, Kentucky 40292, USA
| | - Michael A Menze
- Department of Biology, University of Louisville, Louisville, Kentucky 40292, USA
| | - R Eric Berson
- Department of Chemical Engineering, University of Louisville, Louisville, Kentucky 40292, USA
| | - Paula J Bates
- School of Medicine, University of Louisville, Louisville, Kentucky 40202, USA
| | - Kavitha Yaddanapudi
- Department of Surgery, University of Louisville, Louisville, Kentucky 40202, USA
| | - Jonathan A Kopechek
- Department of Bioengineering, University of Louisville, Louisville, Kentucky 40292, USA
| |
Collapse
|
3
|
Rousou C, Schuurmans CCL, Urtti A, Mastrobattista E, Storm G, Moonen C, Kaarniranta K, Deckers R. Ultrasound and Microbubbles for the Treatment of Ocular Diseases: From Preclinical Research towards Clinical Application. Pharmaceutics 2021; 13:pharmaceutics13111782. [PMID: 34834196 PMCID: PMC8624665 DOI: 10.3390/pharmaceutics13111782] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/08/2021] [Accepted: 10/09/2021] [Indexed: 12/26/2022] Open
Abstract
The unique anatomy of the eye and the presence of various biological barriers make efficacious ocular drug delivery challenging, particularly in the treatment of posterior eye diseases. This review focuses on the combination of ultrasound and microbubbles (USMB) as a minimally invasive method to improve the efficacy and targeting of ocular drug delivery. An extensive overview is given of the in vitro and in vivo studies investigating the mechanical effects of ultrasound-driven microbubbles aiming to: (i) temporarily disrupt the blood–retina barrier in order to enhance the delivery of systemically administered drugs into the eye, (ii) induce intracellular uptake of anticancer drugs and macromolecules and (iii) achieve targeted delivery of genes, for the treatment of ocular malignancies and degenerative diseases. Finally, the safety and tolerability aspects of USMB, essential for the translation of USMB to the clinic, are discussed.
Collapse
Affiliation(s)
- Charis Rousou
- Departments of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Heidelberglaan 8, 3584 CS Utrecht, The Netherlands; (C.C.L.S.); (E.M.); (G.S.)
- Division of Imaging and Oncology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (C.M.); (R.D.)
- Correspondence:
| | - Carl C. L. Schuurmans
- Departments of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Heidelberglaan 8, 3584 CS Utrecht, The Netherlands; (C.C.L.S.); (E.M.); (G.S.)
- Department of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Heidelberglaan 8, 3584 CS Utrecht, The Netherlands
| | - Arto Urtti
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, 70210 Kuopio, Finland;
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014 Helsinki, Finland
- Institute of Chemistry, St. Petersburg State University, Universitetskii Pr. 26, Petrodvorets, 198504 St. Petersburg, Russia
| | - Enrico Mastrobattista
- Departments of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Heidelberglaan 8, 3584 CS Utrecht, The Netherlands; (C.C.L.S.); (E.M.); (G.S.)
| | - Gert Storm
- Departments of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Heidelberglaan 8, 3584 CS Utrecht, The Netherlands; (C.C.L.S.); (E.M.); (G.S.)
- Department of Biomaterials Science and Technology, University of Twente, 7500 AE Enschede, The Netherlands
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Chrit Moonen
- Division of Imaging and Oncology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (C.M.); (R.D.)
| | - Kai Kaarniranta
- Department of Ophthalmology, Kuopio University Hospital, P.O. Box 100, 70029 Kuopio, Finland;
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Roel Deckers
- Division of Imaging and Oncology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (C.M.); (R.D.)
| |
Collapse
|
4
|
Tehrani Fateh S, Moradi L, Kohan E, Hamblin MR, Shiralizadeh Dezfuli A. Comprehensive review on ultrasound-responsive theranostic nanomaterials: mechanisms, structures and medical applications. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2021; 12:808-862. [PMID: 34476167 PMCID: PMC8372309 DOI: 10.3762/bjnano.12.64] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 07/15/2021] [Indexed: 05/03/2023]
Abstract
The field of theranostics has been rapidly growing in recent years and nanotechnology has played a major role in this growth. Nanomaterials can be constructed to respond to a variety of different stimuli which can be internal (enzyme activity, redox potential, pH changes, temperature changes) or external (light, heat, magnetic fields, ultrasound). Theranostic nanomaterials can respond by producing an imaging signal and/or a therapeutic effect, which frequently involves cell death. Since ultrasound (US) is already well established as a clinical imaging modality, it is attractive to combine it with rationally designed nanoparticles for theranostics. The mechanisms of US interactions include cavitation microbubbles (MBs), acoustic droplet vaporization, acoustic radiation force, localized thermal effects, reactive oxygen species generation, sonoluminescence, and sonoporation. These effects can result in the release of encapsulated drugs or genes at the site of interest as well as cell death and considerable image enhancement. The present review discusses US-responsive theranostic nanomaterials under the following categories: MBs, micelles, liposomes (conventional and echogenic), niosomes, nanoemulsions, polymeric nanoparticles, chitosan nanocapsules, dendrimers, hydrogels, nanogels, gold nanoparticles, titania nanostructures, carbon nanostructures, mesoporous silica nanoparticles, fuel-free nano/micromotors.
Collapse
Affiliation(s)
- Sepand Tehrani Fateh
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Lida Moradi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Elmira Kohan
- Department of Science, University of Kurdistan, Kurdistan, Sanandaj, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
| | | |
Collapse
|
5
|
Sanwal R, Joshi K, Ditmans M, Tsai SSH, Lee WL. Ultrasound and Microbubbles for Targeted Drug Delivery to the Lung Endothelium in ARDS: Cellular Mechanisms and Therapeutic Opportunities. Biomedicines 2021; 9:biomedicines9070803. [PMID: 34356867 PMCID: PMC8301318 DOI: 10.3390/biomedicines9070803] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/27/2021] [Accepted: 07/07/2021] [Indexed: 12/16/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is characterized by increased permeability of the alveolar–capillary membrane, a thin barrier composed of adjacent monolayers of alveolar epithelial and lung microvascular endothelial cells. This results in pulmonary edema and severe hypoxemia and is a common cause of death after both viral (e.g., SARS-CoV-2) and bacterial pneumonia. The involvement of the lung in ARDS is notoriously heterogeneous, with consolidated and edematous lung abutting aerated, less injured regions. This makes treatment difficult, as most therapeutic approaches preferentially affect the normal lung regions or are distributed indiscriminately to other organs. In this review, we describe the use of thoracic ultrasound and microbubbles (USMB) to deliver therapeutic cargo (drugs, genes) preferentially to severely injured areas of the lung and in particular to the lung endothelium. While USMB has been explored in other organs, it has been under-appreciated in the treatment of lung injury since ultrasound energy is scattered by air. However, this limitation can be harnessed to direct therapy specifically to severely injured lungs. We explore the cellular mechanisms governing USMB and describe various permutations of cargo administration. Lastly, we discuss both the challenges and potential opportunities presented by USMB in the lung as a tool for both therapy and research.
Collapse
Affiliation(s)
- Rajiv Sanwal
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1T8, Canada; (R.S.); (K.J.); (M.D.); (S.S.H.T.)
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Kushal Joshi
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1T8, Canada; (R.S.); (K.J.); (M.D.); (S.S.H.T.)
- Department of Mechanical and Industrial Engineering, Ryerson University, Toronto, ON M5B 2K3, Canada
- Institute of Biomedical Engineering, Science and Technology (iBEST), Toronto, ON M5B 1T8, Canada
| | - Mihails Ditmans
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1T8, Canada; (R.S.); (K.J.); (M.D.); (S.S.H.T.)
- Biomedical Engineering Graduate Program, Ryerson University, Toronto, ON M5B 2K3, Canada
| | - Scott S. H. Tsai
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1T8, Canada; (R.S.); (K.J.); (M.D.); (S.S.H.T.)
- Department of Mechanical and Industrial Engineering, Ryerson University, Toronto, ON M5B 2K3, Canada
- Institute of Biomedical Engineering, Science and Technology (iBEST), Toronto, ON M5B 1T8, Canada
| | - Warren L. Lee
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1T8, Canada; (R.S.); (K.J.); (M.D.); (S.S.H.T.)
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Mechanical and Industrial Engineering, Ryerson University, Toronto, ON M5B 2K3, Canada
- Institute of Biomedical Engineering, Science and Technology (iBEST), Toronto, ON M5B 1T8, Canada
- Biomedical Engineering Graduate Program, Ryerson University, Toronto, ON M5B 2K3, Canada
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
- Correspondence: ; Tel.: +416-864-6060 (ext. 77655)
| |
Collapse
|
6
|
Maeshige N, Langston PK, Yuan ZM, Kondo H, Fujino H. High-intensity ultrasound irradiation promotes the release of extracellular vesicles from C2C12 myotubes. ULTRASONICS 2021; 110:106243. [PMID: 32961400 DOI: 10.1016/j.ultras.2020.106243] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 08/19/2020] [Accepted: 08/28/2020] [Indexed: 06/11/2023]
Abstract
Skeletal muscle is an important secretory organ in mammals, producing myriad chemical mediators ("myokines") with distinct biological action in different tissues, including anti-inflammatory activity. Extracellular vesicles (EVs) have recently been identified as a mode of myokine transport from muscle, facilitating such anti-inflammatory activity. In this report, we have demonstrated that high-intensity ultrasound (US) strongly induces EV secretion from cultured myotubes without a reduction in cell viability. High-intensity US of 3.0 W/cm2 with 20% duty cycle increased the number of EVs by 2-fold compared to control at 6 h. This effect was specific to EVs in the 100-150 nm size range. Thus, high-intensity US is a novel modality for inducing myocellular EV release and may hold therapeutic value.
Collapse
Affiliation(s)
- Noriaki Maeshige
- Department of Genetics & Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan.
| | - P Kent Langston
- Department of Genetics & Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Immunology, Harvard Medical School and Evergrande Center for Immunologic Diseases, Harvard Medical School, Boston, MA, USA
| | - Zhi-Min Yuan
- Department of Genetics & Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Hiroyo Kondo
- Department of Food Sciences and Nutrition, Nagoya Women's University, Nagoya, Japan
| | - Hidemi Fujino
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| |
Collapse
|
7
|
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: 5.8] [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
|
8
|
Microbubble-facilitated ultrasound pulsation promotes direct α-synuclein gene delivery. Biochem Biophys Res Commun 2019; 517:77-83. [PMID: 31327496 DOI: 10.1016/j.bbrc.2019.07.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 07/05/2019] [Indexed: 01/06/2023]
Abstract
Intra-neuronal α-synuclein (αSNCA) aggregation are the leading cause of dopaminergic neuron degeneration in Parkinson's disease (PD). Most PD patients is linked with αSNCA gene mutations. Gene therapy shows therapeutic potential by packing gene into viral vectors to improve gene expression through stereotactic brain injections. However, through intracranial injection, the gene expression is typically limited with tissue distribution tightly adjacent to the injection track, when expressing therapeutic genes for a wider CNS region is preferable. We use microbubble-facilitated ultrasound pulsations (MB-USP) as a new gene delivering tool to enhance the limit gene delivery of local injection in brain and evaluate the feasibility using αSNCA as model gene. We demonstrate that MB-USP can transfect naked constructs DNA of αSNCA gene into two types of neuron cells and enhance the gene expression. We confirm α-synuclein fusion protein functionality, showing that α-synuclein fusion protein significantly reduce the mitochondrial activity. We show MB-USP improves in vivo gene transfer in the brain with naked construct local injection, significantly enhances α-synuclein expression level to 1.68-fold, and broaden its distribution to 25-fold. In vivo fused α-synuclein protein aggregation is also found in gene-injected mice brains by MB-USP. MB-USP provides an alternative to α-synuclein over expression in vitro and in vivo model for investigation of α-synuclein related PD therapeutic strategies.
Collapse
|
9
|
Fan P, Yang D, Wu J, Yang Y, Guo X, Tu J, Zhang D. Cell-cycle-dependences of membrane permeability and viability observed for HeLa cells undergoing multi-bubble-cell interactions. ULTRASONICS SONOCHEMISTRY 2019; 53:178-186. [PMID: 30642802 DOI: 10.1016/j.ultsonch.2019.01.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 12/30/2018] [Accepted: 01/05/2019] [Indexed: 05/07/2023]
Abstract
Microbubble-mediated sonoporation is a promising strategy for intracellular gene/drug delivery, but the biophysical mechanisms involved in the interactions between microbubbles and cells are not well understood. Here, HeLa cells were synchronized in individual cycle phases, then the cell-cycle-dependences of the membrane permeability and viability of HeLa cells undergoing multi-bubble sonoporation were evaluated using focused ultrasound exposure apparatus coupled passive cavitation detection system. The results indicated that: (1) the microbubble cavitation activity should be independent on cell cycle phases; (2) G1-phase cells with the largest Young's modulus were the most robust against microbubble-mediated sonoporation; (3) G2/M-phase cells exhibited the greatest accumulated FITC uptake with the lowest viability, which should be mainly attributed to the chemical effect of synchronization drugs; and (4) more important, S-phase cells with the lowest stiffness seemed to be the most susceptible to the mechanical effect generated by microbubble cavitation activity, which resulted in the greatest enhancement in sonoporation-facilitated membrane permeabilization without further scarifying their viability. The current findings may benefit ongoing efforts aiming to pursue rational utilization of microbubble-mediated sonoporation in cell-cycle-targeted gene/drug delivery for cancer therapy.
Collapse
Affiliation(s)
- Pengfei Fan
- Key Laboratory of Modern Acoustics (MOE), School of Physics, Nanjing University, Nanjing 210093, China
| | - Dongxin Yang
- Key Laboratory of Modern Acoustics (MOE), School of Physics, Nanjing University, Nanjing 210093, China
| | - Jun Wu
- The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yanye Yang
- Key Laboratory of Modern Acoustics (MOE), School of Physics, Nanjing University, Nanjing 210093, China
| | - Xiasheng Guo
- Key Laboratory of Modern Acoustics (MOE), School of Physics, Nanjing University, Nanjing 210093, China
| | - Juan Tu
- Key Laboratory of Modern Acoustics (MOE), School of Physics, Nanjing University, Nanjing 210093, China.
| | - Dong Zhang
- Key Laboratory of Modern Acoustics (MOE), School of Physics, Nanjing University, Nanjing 210093, China; The State Key Laboratory of Acoustics, Chinese Academy of Science, Beijing 10080, China.
| |
Collapse
|
10
|
Wu X, Liu J, Zhu JJ. Sono-Fenton hybrid process on the inactivation of Microcystis aeruginosa: Extracellular and intracellular oxidation. ULTRASONICS SONOCHEMISTRY 2019; 53:68-76. [PMID: 30600211 DOI: 10.1016/j.ultsonch.2018.12.034] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 12/18/2018] [Accepted: 12/23/2018] [Indexed: 05/26/2023]
Abstract
For the first time, the inactivation of Microcystis aeruginosa using sono-Fenton process at low frequency high intensity (20 kHz, 0.42 W/mL) and high frequency low intensity (800 kHz, 0.07 W/mL) was investigated, respectively. 20 kHz sono-Fenton treatment successfully reduced cyanobacterial cell number from 4.19 × 106 cells/mL to 0.45 × 106 cells/mL within 5 min treatment. Alternatively, efficient performance of 800 kHz sono-Fenton process was observed to decrease Microcystis cell number to 2.33 × 106 cells/mL after 5 min inactivation, with lower energy cost. It was found that powerful 20 kHz sonication induced pore formation on the cell wall, leading to extracellular damage, while 800 kHz irradiation with low intensity triggered intracellular uptake of chemicals, suggesting endocytosis effects. Furthermore, sono-Fenton Processes were found to be affected by the concentrations of Fenton's reagent, and pre-sonication time. Although solo Fenton treatment released microcystins in water, the degradation of microcystin-LR were achieved using 20 and 800 kHz sono-Fenton processes, respectively. The results of this work showed that severe extracellular oxidation is the vital inactivation mechanism of 20 kHz sono-Fenton process, while the internal oxidation caused by intracellularly delivered Fenton reagents is suggested to be the main cause of 800 kHz sono-Fenton inactivation, leading to much lower energy cost. This work provides alternative methods to control harmful cyanobacteria in water towards effective treatment.
Collapse
Affiliation(s)
- Xiaoge Wu
- Environment Science and Engineering College, Yangzhou University, Yangzhou, Jiangsu 225009, China; State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu 210023, China; Jiangsu Collaborative Innovation Center for Solid Organic Waste Resource Utilization, Nanjing 210095, China
| | - Junli Liu
- Environment Science and Engineering College, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Jun-Jie Zhu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu 210023, China.
| |
Collapse
|
11
|
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
|
12
|
Rong N, Zhou H, Liu R, Wang Y, Fan Z. Ultrasound and microbubble mediated plasmid DNA uptake: A fast, global and multi-mechanisms involved process. J Control Release 2018; 273:40-50. [PMID: 29407677 DOI: 10.1016/j.jconrel.2018.01.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Revised: 01/02/2018] [Accepted: 01/16/2018] [Indexed: 11/17/2022]
Abstract
Ultrasound application combined with microbubbles has shown great potential for intracellular gene delivery. However, the fundamental mechanistic question of how plasmid DNA enters the intracellular space mediated by ultrasound and microbubble has not been fully explored and understood. The goal of this study is to unveil the detailed intracellular uptake process of plasmid DNA stimulated by ultrasound and microbubbles, uniquely highlighting the role of microbubbles play in this process. The usage of targeted microbubbles pinpointed the subcellular membrane site, where ultrasound exerted acoustic force onto the cell membrane. With the combination of high-speed video microscopy and 3D confocal fluorescence microscopy, we show the spatiotemporal correlation between the microbubble dynamics and intracellular plasmid DNA distribution. Two ultrasound modes (high pressure short pulse and low pressure long pulse) were chosen to trigger different plasmid DNA uptake routes. We found that reversible cell membrane disruption, induced by high pressure short pulse ultrasound, permitted plasmid DNA passage across cell membrane, but not in an exclusive way. Under both ultrasound modes, with or without cell membrane disruption, global plasmid DNA internalization, even nuclear-localization, was observed immediately post ultrasound application. Our results show that plasmid DNA uptake evoked by localized acoustically excited microbubbles is a fast (<2min), global (not limited to the site where microbubbles were attached), and multi-mechanisms involved process.
Collapse
Affiliation(s)
- Ning Rong
- Department of Biomedical Engineering, Tianjin University, Tianjin 300072, China
| | - Hao Zhou
- College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Ruming Liu
- College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yan Wang
- Department of Biomedical Engineering, Tianjin University, Tianjin 300072, China
| | - Zhenzhen Fan
- Department of Biomedical Engineering, Tianjin University, Tianjin 300072, China; State Key Laboratory of Acoustics, Institute of Acoustics, Chinese Academy of Sciences, Beijing 100190, China.
| |
Collapse
|
13
|
Qin P, Han T, Yu ACH, Xu L. Mechanistic understanding the bioeffects of ultrasound-driven microbubbles to enhance macromolecule delivery. J Control Release 2018; 272:169-181. [PMID: 29305924 DOI: 10.1016/j.jconrel.2018.01.001] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 01/02/2018] [Accepted: 01/03/2018] [Indexed: 12/17/2022]
Abstract
Ultrasound-driven microbubbles can trigger reversible membrane perforation (sonoporation), open interendothelial junctions and stimulate endocytosis, thereby providing a temporary and reversible time-window for the delivery of macromolecules across biological membranes and endothelial barriers. This time-window is related not only to cavitation events, but also to biological regulatory mechanisms. Mechanistic understanding of the interaction between cavitation events and cells and tissues, as well as the subsequent cellular and molecular responses will lead to new design strategies with improved efficacy and minimized side effects. Recent important progress on the spatiotemporal characteristics of sonoporation, cavitation-induced interendothelial gap and endocytosis, and the spatiotemporal bioeffects and the preliminary biological mechanisms in cavitation-enhanced permeability, has been made. On the basis of the summary of this research progress, this Review outlines the underlying bioeffects and the related biological regulatory mechanisms involved in cavitation-enhanced permeability; provides a critical commentary on the future tasks and directions in this field, including developing a standardized methodology to reveal mechanism-based bioeffects in depth, and designing biology-based treatment strategies to improve efficacy and safety. Such mechanistic understanding the bioeffects that contribute to cavitation-enhanced delivery will accelerate the translation of this approach to the clinic.
Collapse
Affiliation(s)
- Peng Qin
- Department of Instrument Science and Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - Tao Han
- Department of Instrument Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Alfred C H Yu
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, ON, Canada
| | - Lin Xu
- National Key Laboratory of Plant Molecular Genetics, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
14
|
Fan P, Zhang Y, Guo X, Cai C, Wang M, Yang D, Li Y, Tu J, Crum LA, Wu J, Zhang D. Cell-cycle-specific Cellular Responses to Sonoporation. Am J Cancer Res 2017; 7:4894-4908. [PMID: 29187912 PMCID: PMC5706108 DOI: 10.7150/thno.20820] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 10/04/2017] [Indexed: 12/22/2022] Open
Abstract
Microbubble-mediated sonoporation has shown its great potential in facilitating intracellular uptake of gene/drugs and other therapeutic agents that are otherwise difficult to enter cells. However, the biophysical mechanisms underlying microbubble-cell interactions remain unclear. Particularly, it is still a major challenge to get a comprehensive understanding of the impact of cell cycle phase on the cellular responses simultaneously occurring in cell membrane and cytoskeleton induced by microbubble sonoporation. Methods: Here, efficient synchronizations were performed to arrest human cervical epithelial carcinoma (HeLa) cells in individual cycle phases. The, topography and stiffness of synchronized cells were examined using atomic force microscopy. The variations in cell membrane permeabilization and cytoskeleton arrangement induced by sonoporation were analyzed simultaneously by a real-time fluorescence imaging system. Results: The results showed that G1-phase cells typically had the largest height and elastic modulus, while S-phase cells were generally the flattest and softest ones. Consequently, the S-Phase was found to be the preferred cycle for instantaneous sonoporation treatment, due to the greatest enhancement of membrane permeability and the fastest cytoskeleton disassembly at the early stage after sonoporation. Conclusion: The current findings may benefit ongoing efforts aiming to pursue rational utilization of microbubble-mediated sonoporation in cell cycle-targeted gene/drug delivery for cancer therapy.
Collapse
|
15
|
Maeshige N, Koga Y, Tanaka M, Aoyama-Ishikawa M, Miyoshi M, Usami M, Fujino H. Low-Intensity Ultrasound Enhances Histone Acetylation and Inhibition of Interleukin 6 Messenger RNA Expression by the Histone Deacetylase Inhibitor Sodium Butyrate in Fibroblasts. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2017; 36:879-885. [PMID: 28195362 DOI: 10.7863/ultra.16.04020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 07/18/2016] [Indexed: 06/06/2023]
Abstract
OBJECTIVES Sodium butyrate, an inhibitor of histone deacetylase, has several therapeutic actions, including anti-inflammation. These actions depend on the concentration of sodium butyrate. In addition, lower concentrations have shown no effect on inflammation. Sonoporation by ultrasound can modify the permeability of the cell plasma membrane. Thus, the effects of sodium butyrate may be enhanced by the ultrasonic acoustics. Therefore, the facilitative effects of low-intensity ultrasound on histone acetylation and interleukin 6 (IL-6) regulation by sodium butyrate were investigated in this study. METHODS Human dermal fibroblasts were treated with 1-mM sodium butyrate for 3 hours with 20 minutes of 0.1-W/cm2 pulsed or continuous ultrasound irradiation at the beginning of the sodium butyrate treatments. RESULTS The combination of treatments with sodium butyrate and ultrasound significantly increased histone acetylation in fibroblasts (P < .05), whereas sodium butyrate could not increase histone acetylation. In addition, this combined treatment significantly suppressed the IL-6 messenger RNA expression level with lipopolysaccharide stimulation for 1 hour (P < .05). Meanwhile, the treatment with sodium butyrate alone could not suppress IL-6 messenger RNA expression in fibroblasts. These effects were achieved with both 20% pulsed and continuous ultrasound but not observed with ultrasound treatment alone. CONCLUSIONS These results suggest that low-intensity ultrasound treatment promotes the physiologic actions of sodium butyrate as a histone deacetylase inhibitor.
Collapse
Affiliation(s)
- Noriaki Maeshige
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Yuka Koga
- Division of Nutrition and Metabolism , Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Masayuki Tanaka
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Michiko Aoyama-Ishikawa
- Division of Nutrition and Metabolism , Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Makoto Miyoshi
- Division of Nutrition and Metabolism , Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Makoto Usami
- Division of Nutrition and Metabolism , Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Hidemi Fujino
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| |
Collapse
|
16
|
Maciulevičius M, Tamošiūnas M, Jakštys B, Jurkonis R, Venslauskas MS, Šatkauskas S. Investigation of Microbubble Cavitation-Induced Calcein Release from Cells In Vitro. ULTRASOUND IN MEDICINE & BIOLOGY 2016; 42:2990-3000. [PMID: 27637933 DOI: 10.1016/j.ultrasmedbio.2016.08.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 07/13/2016] [Accepted: 08/02/2016] [Indexed: 06/06/2023]
Abstract
In the present study, microbubble (MB) cavitation signal analysis was performed together with calcein release evaluation in both pressure and exposure duration domains of the acoustic field. A passive cavitation detection system was used to simultaneously measure MB scattering and attenuation signals for subsequent extraction efficiency relative to MB cavitation activity. The results indicate that the decrease in the efficiency of extraction of calcein molecules from Chinese hamster ovary cells, as well as cell viability, is associated with MB cavitation activity and can be accurately predicted using inertial cavitation doses up to 0.18 V × s (R2 > 0.9, p < 0.0001). No decrease in additional calcein release or cell viability was observed after complete MB sonodestruction was achieved. This indicates that the optimal exposure duration within which maximal sono-extraction efficiency is obtained coincides with the time necessary to achieve complete MB destruction. These results illustrate the importance of MB inertial cavitation in the sono-extraction process. To our knowledge, this study is the first to (i) investigate small molecule extraction from cells via sonoporation and (ii) relate the extraction process to the quantitative characteristics of MB cavitation acoustic spectra.
Collapse
Affiliation(s)
| | | | | | - Rytis Jurkonis
- Biomedical Engineering Institute, Kaunas University of Technology, Kaunas, Lithuania
| | | | - Saulius Šatkauskas
- Biophysical Research Group, Vytautas Magnus University, Kaunas, Lithuania.
| |
Collapse
|
17
|
Liu Y, Yan J, Santangelo PJ, Prausnitz MR. DNA uptake, intracellular trafficking and gene transfection after ultrasound exposure. J Control Release 2016; 234:1-9. [PMID: 27165808 DOI: 10.1016/j.jconrel.2016.05.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 04/26/2016] [Accepted: 05/06/2016] [Indexed: 11/17/2022]
Abstract
Ultrasound has been studied as a promising tool for intracellular gene delivery. In this work, we studied gene transfection of a human prostate cancer cell line exposed to megahertz pulsed ultrasound in the presence of contrast agent and assessed the efficiency of fluorescently labelled DNA delivery into cell nuclei, which is necessary for gene transfection. At the sonication conditions studied, ~30% of cells showed DNA uptake 30min after sonication, but that fraction decreased over time to ~10% of cells after 24h. Most cells containing DNA had DNA in their nuclei, but the amount varied significantly. Transfection efficiency peaked at ~10% at 8h post sonication. Among those cells containing DNA, ~30% of DNA was localized in the cell nuclei, ~30% was in autophagosomes/autophagolysosomes and the remainder was "free" in the cytoplasm 30min after sonication. At later times up to 24h, ~30% of DNA continued to be found in the nuclei and most or all of the rest of the DNA was in autophagosomes/autophagolysosomes. These results demonstrate that ultrasound can deliver DNA into cell nuclei shortly after sonication and that the rest of the DNA can be cleared by autophagosomes/autophagolysosomes.
Collapse
Affiliation(s)
- Ying Liu
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332-0100, USA
| | - Jing Yan
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332-0100, USA
| | - Philip J Santangelo
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory University, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Mark R Prausnitz
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332-0100, USA; Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory University, Georgia Institute of Technology, Atlanta, GA 30332, USA.
| |
Collapse
|
18
|
Lee HJ, Yoon YI, Bae YJ. Theragnostic ultrasound using microbubbles in the treatment of prostate cancer. Ultrasonography 2016; 35:309-17. [PMID: 27197842 PMCID: PMC5040139 DOI: 10.14366/usg.16006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 04/14/2016] [Accepted: 04/14/2016] [Indexed: 12/19/2022] Open
Abstract
The use of gas-filled microbubbles in perfusion monitoring as intravascular ultrasound contrast agents has recently become more common. Additionally, microbubbles are employed as carriers of pharmaceutical substances or genes. Microbubbles have great potential to improve the delivery of therapeutic materials into cells and to modify vascular permeability, causing increased extravasation of drugs and drug carriers. Prostate cancer is the most common neoplasm in Europe and America, with an incidence twice to three times that of lung and colorectal cancer. Its incidence is still rising in Asian countries, including Japan and Korea. In this review, we present current strategies regarding the synthesis of microbubbles with targeted ligands on their surfaces, with a focus on prostate cancer.
Collapse
Affiliation(s)
- Hak Jong Lee
- Department of Radiology, Seoul National University Bundang Hospital, Seongnam, Korea.,Department of Radiology, Seoul National University College of Medicine, Seoul, Korea.,Program in Nanoscience and Technology, Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| | - Young Il Yoon
- Department of Radiology, Seoul National University Bundang Hospital, Seongnam, Korea.,Program in Nanoscience and Technology, Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| | - Yun Jung Bae
- Department of Radiology, Seoul National University Bundang Hospital, Seongnam, Korea.,Department of Radiology, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
19
|
Bouakaz A, Zeghimi A, Doinikov AA. Sonoporation: Concept and Mechanisms. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 880:175-89. [PMID: 26486338 DOI: 10.1007/978-3-319-22536-4_10] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Contrast agents for ultrasound are now routinely used for diagnosis and imaging. In recent years, new promising possibilities for targeted drug delivery have been proposed that can be realized by using the microbubble composing ultrasound contrast agents (UCAs). The microbubbles can carry drugs and selectively adhere to specific sites in the human body. This capability, in combination with the effect known as sonoporation, provides great possibilities for localized drug delivery. Sonoporation is a process in which ultrasonically activated UCAs, pulsating nearby biological barriers (cell membrane or endothelial layer), increase their permeability and thereby enhance the extravasation of external substances. In this way drugs and genes can be delivered inside individual cells without serious consequences for the cell viability. Sonoporation has been validated both in-vitro using cell cultures and in-vivo in preclinical studies. However, today, the mechanisms by which molecules cross the biological barriers remain unrevealed despite a number of proposed theories. This chapter will provide a survey of the current studies on various hypotheses regarding the routes by which drugs are incorporated into cells or across the endothelial layer and possible associated microbubble acoustic phenomena.
Collapse
Affiliation(s)
- Ayache Bouakaz
- Inserm Imaging and Ultrasound, INSERM U930, Imagerie et Cerveau, Université François-Rabelais de Tours, Tours, France.
| | - Aya Zeghimi
- Inserm Imaging and Ultrasound, INSERM U930, Imagerie et Cerveau, Université François-Rabelais de Tours, Tours, France
| | - Alexander A Doinikov
- Inserm Imaging and Ultrasound, INSERM U930, Imagerie et Cerveau, Université François-Rabelais de Tours, Tours, France
| |
Collapse
|
20
|
Nande R, Howard CM, Claudio PP. Ultrasound-mediated oncolytic virus delivery and uptake for increased therapeutic efficacy: state of art. Oncolytic Virother 2015; 4:193-205. [PMID: 27512682 PMCID: PMC4918399 DOI: 10.2147/ov.s66097] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The field of ultrasound (US) has changed significantly from medical imaging and diagnosis to treatment strategies. US contrast agents or microbubbles (MB) are currently being used as potential carriers for chemodrugs, small molecules, nucleic acids, small interfering ribonucleic acid, proteins, adenoviruses, and oncolytic viruses. Oncolytic viruses can selectively replicate within and destroy a cancer cell, thus making them a powerful therapeutic in treating late-stage or metastatic cancer. These viruses have been shown to have robust activity in clinical trials when injected directly into tumor nodules. However limitations in oncolytic virus’ effectiveness and its delivery approach have warranted exploration of ultrasound-mediated delivery. Gene therapy bearing adenoviruses or oncolytic viruses can be coupled with MBs and injected intravenously. Following application of US energy to the target region, the MBs cavitate, and the resulting shock wave enhances drug, gene, or adenovirus uptake. Though the underlying mechanism is yet to be fully understood, there is evidence to suggest that mechanical pore formation of cellular membranes allows for the temporary uptake of drugs. This delivery method circumvents the limitations due to stimulation of the immune system that prevented intravenous administration of viruses. This review provides insight into this intriguing new frontier on the delivery of oncolytic viruses to tumor sites.
Collapse
Affiliation(s)
- Rounak Nande
- Department of Biochemistry and Microbiology, Marshall University School of Medicine, Huntington, WV, USA
| | - Candace M Howard
- Department of Radiology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Pier Paolo Claudio
- Department of BioMolecular Sciences and National Center for Natural Products Research, School of Pharmacy, University of Mississippi, MS, USA; Department of Radiation Oncology, University of Mississippi Medical Center, Jackson, MS, USA
| |
Collapse
|
21
|
Delalande A, Leduc C, Midoux P, Postema M, Pichon C. Efficient Gene Delivery by Sonoporation Is Associated with Microbubble Entry into Cells and the Clathrin-Dependent Endocytosis Pathway. ULTRASOUND IN MEDICINE & BIOLOGY 2015; 41:1913-1926. [PMID: 25929996 DOI: 10.1016/j.ultrasmedbio.2015.03.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 03/06/2015] [Accepted: 03/07/2015] [Indexed: 06/04/2023]
Abstract
Microbubble oscillation at specific ultrasound settings leads to permeabilization of surrounding cells. This phenomenon, referred to as sonoporation, allows for the in vitro and in vivo delivery of extracellular molecules, including plasmid DNA. To date, the biological and physical mechanisms underlying this phenomenon are not fully understood. The aim of this study was to investigate the interactions between microbubbles and cells, as well as the intracellular routing of plasmid DNA and microbubbles, during and after sonoporation. High-speed imaging and fluorescence confocal microscopy of HeLa cells stably expressing enhanced green fluorescent protein fused with markers of cellular compartments were used for this investigation. Soft-shelled microbubbles were observed to enter cells during sonoporation using experimental parameters that led to optimal gene transfer. They interacted with the plasma membrane in a specific area stained with fluorescent cholera subunit B, a marker of lipid rafts. This process was not observed with hard-shelled microbubbles, which were not efficient in gene delivery under our conditions. The plasmid DNA was delivered to late endosomes after 3 h post-sonoporation, and a few were found in the nucleus after 6 h. Gene transfer efficacy was greatly inhibited when cells were treated with chlorpromazine, an inhibitor of the clathrin-dependent endocytosis pathway. In contrast, no significant alteration was observed when cells were treated with filipin III or genistein, both inhibitors of the caveolin-dependent pathway. This study emphasizes that microbubble-cell interactions do not occur randomly during sonoporation; microbubble penetration inside cells affects the efficacy of gene transfer at specific ultrasound settings; and plasmid DNA uptake is an active mechanism that involves the clathrin-dependent pathway.
Collapse
Affiliation(s)
| | - Chloé Leduc
- Centre de Biophysique Moléculaire, CNRS UPR4301, Orléans, France
| | - Patrick Midoux
- Centre de Biophysique Moléculaire, CNRS UPR4301, Orléans, France
| | - Michiel Postema
- Department of Physics and Technology, University of Bergen, Bergen, Norway; Department of Physics, University of the Witwatersrand, Johannesburg, South Africa
| | - Chantal Pichon
- Centre de Biophysique Moléculaire, CNRS UPR4301, Orléans, France
| |
Collapse
|
22
|
Derieppe M, Rojek K, Escoffre JM, de Senneville BD, Moonen C, Bos C. Recruitment of endocytosis in sonopermeabilization-mediated drug delivery: a real-time study. Phys Biol 2015; 12:046010. [PMID: 26118644 DOI: 10.1088/1478-3975/12/4/046010] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Microbubbles (MBs) in combination with ultrasound (US) can enhance cell membrane permeability, and have the potential to facilitate the cellular uptake of hydrophilic molecules. However, the exact mechanism behind US- and MB-mediated intracellular delivery still remains to be fully understood. Among the proposed mechanisms are formation of transient pores and endocytosis stimulation. In our study, we investigated whether endocytosis is involved in US- and MB-mediated delivery of small molecules. Dynamic fluorescence microscopy was used to investigate the effects of endocytosis inhibitors on the pharmacokinetic parameters of US- and MB-mediated uptake of SYTOX Green, a 600 Da hydrophilic model drug. C6 rat glioma cells, together with SonoVue(®) MBs, were exposed to 1.4 MHz US waves at 0.2 MPa peak-negative pressure. Collection of the signal intensity in each individual nucleus was monitored during and after US exposure by a fibered confocal fluorescence microscope designed for real-time imaging. Exposed to US waves, C6 cells pretreated with chlorpromazine, an inhibitor of clathrin-mediated endocytosis, showed up to a 2.5-fold significant increase of the uptake time constant, and a 1.1-fold increase with genistein, an inhibitor of caveolae-mediated endocytosis. Both inhibitors slowed down the US-mediated uptake of SYTOX Green. With C6 cells and our experimental settings, these quantitative data indicate that endocytosis plays a role in sonopermeabilization-mediated delivery of small molecules with a more predominant contribution of clathrin-mediated endocytosis.
Collapse
Affiliation(s)
- Marc Derieppe
- Imaging Division, University Medical Center Utrecht, Utrecht, Netherlands
| | | | | | | | | | | |
Collapse
|
23
|
Lentacker I, De Cock I, Deckers R, De Smedt SC, Moonen CTW. Understanding ultrasound induced sonoporation: definitions and underlying mechanisms. Adv Drug Deliv Rev 2014; 72:49-64. [PMID: 24270006 DOI: 10.1016/j.addr.2013.11.008] [Citation(s) in RCA: 511] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 11/13/2013] [Indexed: 01/01/2023]
Abstract
In the past two decades, research has underlined the potential of ultrasound and microbubbles to enhance drug delivery. However, there is less consensus on the biophysical and biological mechanisms leading to this enhanced delivery. Sonoporation, i.e. the formation of temporary pores in the cell membrane, as well as enhanced endocytosis is reported. Because of the variety of ultrasound settings used and corresponding microbubble behavior, a clear overview is missing. Therefore, in this review, the mechanisms contributing to sonoporation are categorized according to three ultrasound settings: i) low intensity ultrasound leading to stable cavitation of microbubbles, ii) high intensity ultrasound leading to inertial cavitation with microbubble collapse, and iii) ultrasound application in the absence of microbubbles. Using low intensity ultrasound, the endocytotic uptake of several drugs could be stimulated, while short but intense ultrasound pulses can be applied to induce pore formation and the direct cytoplasmic uptake of drugs. Ultrasound intensities may be adapted to create pore sizes correlating with drug size. Small molecules are able to diffuse passively through small pores created by low intensity ultrasound treatment. However, delivery of larger drugs such as nanoparticles and gene complexes, will require higher ultrasound intensities in order to allow direct cytoplasmic entry.
Collapse
Affiliation(s)
- I Lentacker
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium
| | - I De Cock
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium
| | - R Deckers
- Imaging Division, University Medical Center Utrecht, PO Box 85500, 3508 GA Utrecht, The Netherlands
| | - S C De Smedt
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium.
| | - C T W Moonen
- Imaging Division, University Medical Center Utrecht, PO Box 85500, 3508 GA Utrecht, The Netherlands
| |
Collapse
|
24
|
Tsai WB, Lai HY, Lee JL, Lo CW, Chen WS. Enhancement of the cytotoxicity and selectivity of doxorubicin to hepatoma cells by synergistic combination of galactose-decorated γ-poly(glutamic acid) nanoparticles and low-intensity ultrasound. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2014; 30:5510-5517. [PMID: 24754730 DOI: 10.1021/la500352g] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Specific drug delivery to solid tumors remains one of the challenges in cancer therapy. The aim of this study was to combine three drug-targeting strategies, polymer-drug conjugate, ligand presentation and ultrasound treatment, to enhance the efficacy and selectivity of doxorubicin (DXR) to hepatoma cells. The conjugation of DXR to γ-poly(glutamic acids) (γ-PGA) decreased the cytotoxicity of DXR, while the conjugation of galactosamine (Gal) to the γ-PGA-DXR conjugate restored the cytotoxic efficacy of DXR on hepatoma cells due to increased uptake of DXR. Furthermore, low-intensity ultrasound treatment increased the cell-killing ability of γ-PGA-DXR conjugates by 20%. The in vitro results showed the potential of the γ-PGA-DXR-Gal conjugate for future clinical applications.
Collapse
Affiliation(s)
- Wei-Bor Tsai
- Department of Chemical Engineering, National Taiwan University , Number 1, Section 4, Roosevelt Road, Taipei 106, Taiwan
| | | | | | | | | |
Collapse
|
25
|
Juffermans LJM, Meijering BDM, Henning RH, Deelman LE. Ultrasound and microbubble-targeted delivery of small interfering RNA into primary endothelial cells is more effective than delivery of plasmid DNA. ULTRASOUND IN MEDICINE & BIOLOGY 2014; 40:532-540. [PMID: 24361223 DOI: 10.1016/j.ultrasmedbio.2013.09.031] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Revised: 09/27/2013] [Accepted: 09/30/2013] [Indexed: 06/03/2023]
Abstract
Ultrasound and microbubble-targeted delivery (UMTD) is a promising non-viral technique for genetic-based therapy. We found that UMTD of small interfering RNA (siRNA) is more effective than delivery of plasmid DNA (pDNA). UMTD (1 MHz, 0.22 MPa) of fluorescently labeled siRNA resulted in 97.9 ± 1.5% transfected cells, with siRNA localized homogenously in the cytoplasm directly after ultrasound exposure. UMTD of fluorescently labeled pDNA resulted in only 43.0 ± 4.2% transfected cells, with localization mainly in vesicular structures, co-localizing with endocytosis markers clathrin and caveolin. Delivery of siRNA against GAPDH (glyceraldehyde-3-phosphate dehydrogenase) effectively decreased protein levels to 24.3 ± 7.9% of non-treated controls (p < 0.01). In contrast, 24 h after delivery of pDNA encoding GAPDH, no increase in protein levels was detected. Transfection efficiency, verified with red fluorescently labeled pDNA encoding enhanced green fluorescent protein, revealed that of the transfected cells, only 2.0 ± 0.7% expressed the transgene. In conclusion, the difference in localization between siRNA and pDNA after UMTD is an important determinant of the effectiveness of these genetic-based technologies.
Collapse
Affiliation(s)
- Lynda J M Juffermans
- Departments of Physiology and Cardiology, VU University Medical Center, Amsterdam, The Netherlands; Department of Clinical Pharmacology, Groningen Institute for Drug Exploration (GUIDE), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - Bernadet D M Meijering
- Department of Clinical Pharmacology, Groningen Institute for Drug Exploration (GUIDE), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands
| | - Robert H Henning
- Department of Clinical Pharmacology, Groningen Institute for Drug Exploration (GUIDE), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands
| | - Leo E Deelman
- Department of Clinical Pharmacology, Groningen Institute for Drug Exploration (GUIDE), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
26
|
Jin L, Li F, Wang H, Li Y, Wei F, Du L. Ultrasound‑targeted microbubble destruction enhances gene transduction of adeno-associated virus in a less-permissive cell type, NIH/3T3. Mol Med Rep 2013; 8:320-6. [PMID: 23817930 PMCID: PMC3776706 DOI: 10.3892/mmr.2013.1560] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 06/13/2013] [Indexed: 12/17/2022] Open
Abstract
Adeno‑associated virus (AAV) is a common vector utilized in gene therapy. The NIH/3T3 cell line, which is a potential induced pluripotent stem (iPS) cell type, was identified to be a less-permissive cell type to AAV due to its defective endosomal processing. Ultrasound‑targeted microbubble destruction (UTMD) enhanced the gene transduction of AAV in permissive cells. However, there are no data concerning UTMD enhancement in less-permissive cells, and the exact mechanism of UTMD enhancement in cellular uptake is unclear. Greater knowledge concerning the rate-limiting steps in NIH/3T3 cells would aid in the elucidation of the mechanism of UTMD enhancement in the gene transduction of AAV. In the present study, UTMD enhanced the gene transduction of AAV in NIH/3T3 cells, suggesting that UTMD‑enhanced AAV‑mediated gene transduction may be beneficial for gene therapy in iPS cells. The dose dependence of UTMD enhancement indicated that mechanisms other than sonoporation were involved in the cellular uptake of AAV. However, UTMD did not greatly increase the gene transduction of AAV in NIH/3T3 cells. Additionally, the similar degree of enhancement in the two cell types resulted in no correlation between UTMD and endosomal processing. Future studies on UTMD‑mediated AAV transduction in other non- or less‑permissive cell types may aid in elucidating the exact mechanism of UTMD enhancement in cellular uptake.
Collapse
Affiliation(s)
- Lifang Jin
- Department of Ultrasound, Shanghai First People's Hospital, Shanghai Jiao Tong University, Shanghai 200080, P.R. China
| | | | | | | | | | | |
Collapse
|
27
|
Burgess A, Hynynen K. Noninvasive and targeted drug delivery to the brain using focused ultrasound. ACS Chem Neurosci 2013; 4:519-26. [PMID: 23379618 PMCID: PMC3629738 DOI: 10.1021/cn300191b] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Accepted: 01/21/2013] [Indexed: 01/27/2023] Open
Abstract
Brain diseases are notoriously difficult to treat due to the presence of the blood-brain barrier (BBB). Here, we review the development of focused ultrasound (FUS) as a noninvasive method for BBB disruption, aiding in drug delivery to the brain. FUS can be applied through the skull to a targeted region in the brain. When combined with microbubbles, FUS causes localized and reversible disruption of the BBB. The cellular mechanisms of BBB disruption are presented. Several therapeutic agents have been delivered to the brain resulting in significant improvements in pathology in models of glioblastoma and Alzheimer's disease. The requirements for clinical translation of FUS will be discussed.
Collapse
Affiliation(s)
- Alison Burgess
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Kullervo Hynynen
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Medical Biophysics and Institute
of Biomaterials & Biomedical Engineering (IBBME), University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
28
|
Delalande A, Kotopoulis S, Postema M, Midoux P, Pichon C. Sonoporation: mechanistic insights and ongoing challenges for gene transfer. Gene 2013; 525:191-9. [PMID: 23566843 DOI: 10.1016/j.gene.2013.03.095] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 02/27/2013] [Accepted: 03/07/2013] [Indexed: 11/29/2022]
Abstract
Microbubbles first developed as ultrasound contrast agents have been used to assist ultrasound for cellular drug and gene delivery. Their oscillation behavior during ultrasound exposure leads to transient membrane permeability of surrounding cells, facilitating targeted local delivery. The increased cell uptake of extracellular compounds by ultrasound in the presence of microbubbles is attributed to a phenomenon called sonoporation. In this review, we summarize current state of the art concerning microbubble-cell interactions and cellular effects leading to sonoporation and its application for gene delivery. Optimization of sonoporation protocol and composition of microbubbles for gene delivery are discussed.
Collapse
|
29
|
Ultrasound and microbubble-assisted gene delivery: recent advances and ongoing challenges. Ther Deliv 2012; 3:1199-215. [PMID: 23116012 DOI: 10.4155/tde.12.100] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Having first been developed for ultrasound imaging, nowadays, microbubbles are proposed as tools for ultrasound-assisted gene delivery, too. Their behavior during ultrasound exposure causes transient membrane permeability of surrounding cells, facilitating targeted local delivery. The increased cell uptake of extracellular compounds by ultrasound in the presence of microbubbles is attributed to a phenomenon called sonoporation. Sonoporation has been successfully applied to deliver nucleic acids in vitro and in vivo in a variety of therapeutic applications. However, the biological and physical mechanisms of sonoporation are still not fully understood. In this review, we discuss recent data concerning microbubble--cell interactions leading to sonoporation and we report on the progress in ultrasound-assisted therapeutic gene delivery in different organs. In addition, we outline ongoing challenges of this novel delivery method for its clinical use.
Collapse
|
30
|
Lee JL, Lo CW, Ka SM, Chen A, Chen WS. Prolonging the expression duration of ultrasound-mediated gene transfection using PEI nanoparticles. J Control Release 2012; 160:64-71. [DOI: 10.1016/j.jconrel.2012.03.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2011] [Revised: 02/18/2012] [Accepted: 03/09/2012] [Indexed: 02/07/2023]
|