1
|
Chen J, Geng J, Li Y, Xia P, Li X, Wang F, Chen D, Wang M, Wang H. Investigation of incipient cavitation in various liquids based on PIV quantification and numerical simulations. Sci Rep 2025; 15:9390. [PMID: 40102570 PMCID: PMC11920098 DOI: 10.1038/s41598-025-93746-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 03/10/2025] [Indexed: 03/20/2025] Open
Abstract
This work experimentally and analytically investigated the incipient cavitation behavior in four liquids with different physical properties: ethanol, de-ionized water, glycerine, and aluminum melt under ultrasonic irradiation close to the cavitation threshold with a frequency of 20 kHz. To identify the cavitation structure development and bubble motion of different liquids, cavitation structure under condition close to the cavitation threshold was in-situ observed via high-speed photography for optically transparent liquids and synchrotron radiation X-ray radiography technology for aluminum melt. The dynamic process of the cavitation bubble was numerically simulated on the bubble wall motion. Bubble characteristics were analyzed by more accurate relevant dimensionless quantities comparison obtained from the translational maximum bubble velocity measured by Particle Image Velocimetry (PIV). Based on the simulation results through multi-technology combination methods and dynamic simulation, the incipient cavitation characteristics of various liquid bulks in experimental conditions were estimated and compared. The insights gained from this study are valuable for improving the design and optimization of industrial processes involving cavitation, such as ultrasonic degassing, ultrasonic-assisted metal casting, and material processing. Understanding these cavitation characteristics can lead to more efficient and controlled applications in these fields and help in identifying more suitable transparent media for simulating the cavitation behavior of metal melts.
Collapse
Affiliation(s)
- Jian Chen
- State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai, 200240, China
| | - Jiwei Geng
- State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai, 200240, China
- Anhui Provincial Industrial Generic Technology Research Center for Alumics Materials, Huaibei Normal University, Huaibei, 235000, China
| | - Yugang Li
- State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai, 200240, China
- Anhui Provincial Industrial Generic Technology Research Center for Alumics Materials, Huaibei Normal University, Huaibei, 235000, China
| | - Peikang Xia
- State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai, 200240, China
- Anhui Provincial Engineering Research Center of Aluminium Matrix Composites, Huaibei, 235000, China
| | - Xianfeng Li
- State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai, 200240, China
- Anhui Provincial Industrial Generic Technology Research Center for Alumics Materials, Huaibei Normal University, Huaibei, 235000, China
- Anhui Provincial Engineering Research Center of Aluminium Matrix Composites, Huaibei, 235000, China
| | - Fangming Wang
- CNPC Engineering Technology R&d Company Limited, Beijing, 102206, China
| | - Dong Chen
- State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai, 200240, China.
- Anhui Provincial Industrial Generic Technology Research Center for Alumics Materials, Huaibei Normal University, Huaibei, 235000, China.
- Anhui Provincial Engineering Research Center of Aluminium Matrix Composites, Huaibei, 235000, China.
| | - Mingliang Wang
- State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai, 200240, China.
- Anhui Provincial Industrial Generic Technology Research Center for Alumics Materials, Huaibei Normal University, Huaibei, 235000, China.
- Anhui Provincial Engineering Research Center of Aluminium Matrix Composites, Huaibei, 235000, China.
| | - Haowei Wang
- State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai, 200240, China
- Anhui Provincial Industrial Generic Technology Research Center for Alumics Materials, Huaibei Normal University, Huaibei, 235000, China
- Anhui Provincial Engineering Research Center of Aluminium Matrix Composites, Huaibei, 235000, China
| |
Collapse
|
2
|
Berger S, Zeyn Y, Wagner E, Bros M. New insights for the development of efficient DNA vaccines. Microb Biotechnol 2024; 17:e70053. [PMID: 39545748 PMCID: PMC11565620 DOI: 10.1111/1751-7915.70053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 10/29/2024] [Indexed: 11/17/2024] Open
Abstract
Despite the great potential of DNA vaccines for a broad range of applications, ranging from prevention of infections, over treatment of autoimmune and allergic diseases to cancer immunotherapies, the implementation of such therapies for clinical treatment is far behind the expectations up to now. The main reason is the poor immunogenicity of DNA vaccines in humans. Consequently, the improvement of the performance of DNA vaccines in vivo is required. This mini-review provides an overview of the current state of DNA vaccines and the various strategies to enhance the immunogenic potential of DNA vaccines, including (i) the optimization of the DNA construct itself regarding size, nuclear transfer and transcriptional regulation; (ii) the use of appropriate adjuvants; and (iii) improved delivery, for example, by careful choice of the administration route, physical methods such as electroporation and nanomaterials that may allow cell type-specific targeting. Moreover, combining nanoformulated DNA vaccines with other immunotherapies and prime-boost strategies may help to enhance success of treatment.
Collapse
Affiliation(s)
- Simone Berger
- Pharmaceutical Biotechnology, Department of Pharmacy, Center for NanoScienceLudwig‐Maximilians‐Universität (LMU) MunichMunichGermany
| | - Yanira Zeyn
- Department of DermatologyUniversity Medical Center of the Johannes Gutenberg University (JGU) MainzMainzGermany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Center for NanoScienceLudwig‐Maximilians‐Universität (LMU) MunichMunichGermany
| | - Matthias Bros
- Department of DermatologyUniversity Medical Center of the Johannes Gutenberg University (JGU) MainzMainzGermany
| |
Collapse
|
3
|
Li X, Hu Y, Zhang X, Shi X, Parak WJ, Pich A. Transvascular transport of nanocarriers for tumor delivery. Nat Commun 2024; 15:8172. [PMID: 39289401 PMCID: PMC11408679 DOI: 10.1038/s41467-024-52416-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 09/05/2024] [Indexed: 09/19/2024] Open
Abstract
Nanocarriers (NCs) play a crucial role in delivering theranostic agents to tumors, making them a pivotal focus of research. However, the persistently low delivery efficiency of engineered NCs has been a significant challenge in the advancement of nanomedicine, stirring considerable debate. Transvascular transport is a critical pathway for NC delivery from vessels to tumors, yet a comprehensive understanding of the interactions between NCs and vascular systems remains elusive. In recent years, considerable efforts have been invested in elucidating the transvascular transport mechanisms of NCs, leading to promising advancements in tumor delivery and theranostics. In this context, we highlight various delivery mechanisms, including the enhanced permeability and retention effect, cooperative immune-driven effect, active transcytosis, and cell/bacteria-mediated delivery. Furthermore, we explore corresponding strategies aimed at enhancing transvascular transport of NCs for efficient tumor delivery. These approaches offer intriguing solutions spanning physicochemical, biological, and pharmacological domains to improve delivery and therapeutic outcomes. Additionally, we propose a forward-looking delivery framework that relies on advanced tumor/vessel models, high-throughput NC libraries, nano-bio interaction datasets, and artificial intelligence, which aims to guide the design of next-generation carriers and implementation strategies for optimized delivery.
Collapse
Affiliation(s)
- Xin Li
- DWI-Leibniz-Institute for Interactive Materials, Aachen, 52056, Germany
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Aachen, 52074, Germany
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, Shanghai, 201804, China
| | - Yong Hu
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, Shanghai, 201804, China
| | - Xingcai Zhang
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA.
| | - Xiangyang Shi
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Wolfgang J Parak
- Center for Hybrid Nanostructures (CHyN), University of Hamburg, Hamburg, 20607, Germany.
| | - Andrij Pich
- DWI-Leibniz-Institute for Interactive Materials, Aachen, 52056, Germany.
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Aachen, 52074, Germany.
- Aachen Maastricht Institute for Biobased Materials, Maastricht University, RD Geleen, 6167, The Netherlands.
| |
Collapse
|
4
|
Shakya G, Cattaneo M, Guerriero G, Prasanna A, Fiorini S, Supponen O. Ultrasound-responsive microbubbles and nanodroplets: A pathway to targeted drug delivery. Adv Drug Deliv Rev 2024; 206:115178. [PMID: 38199257 DOI: 10.1016/j.addr.2023.115178] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/21/2023] [Accepted: 12/31/2023] [Indexed: 01/12/2024]
Abstract
Ultrasound-responsive agents have shown great potential as targeted drug delivery agents, effectively augmenting cell permeability and facilitating drug absorption. This review focuses on two specific agents, microbubbles and nanodroplets, and provides a sequential overview of their drug delivery process. Particular emphasis is given to the mechanical response of the agents under ultrasound, and the subsequent physical and biological effects on the cells. Finally, the state-of-the-art in their pre-clinical and clinical implementation are discussed. Throughout the review, major challenges that need to be overcome in order to accelerate their clinical translation are highlighted.
Collapse
Affiliation(s)
- Gazendra Shakya
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland
| | - Marco Cattaneo
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland
| | - Giulia Guerriero
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland
| | - Anunay Prasanna
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland
| | - Samuele Fiorini
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland
| | - Outi Supponen
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland.
| |
Collapse
|
5
|
Rajora MA, Dhaliwal A, Zheng M, Choi V, Overchuk M, Lou JWH, Pellow C, Goertz D, Chen J, Zheng G. Quantitative Pharmacokinetics Reveal Impact of Lipid Composition on Microbubble and Nanoprogeny Shell Fate. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304453. [PMID: 38032129 PMCID: PMC10811482 DOI: 10.1002/advs.202304453] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/19/2023] [Indexed: 12/01/2023]
Abstract
Microbubble-enabled focused ultrasound (MB-FUS) has revolutionized nano and molecular drug delivery capabilities. Yet, the absence of longitudinal, systematic, quantitative studies of microbubble shell pharmacokinetics hinders progress within the MB-FUS field. Microbubble radiolabeling challenges contribute to this void. This barrier is overcome by developing a one-pot, purification-free copper chelation protocol able to stably radiolabel diverse porphyrin-lipid-containing Definity® analogues (pDefs) with >95% efficiency while maintaining microbubble physicochemical properties. Five tri-modal (ultrasound-, positron emission tomography (PET)-, and fluorescent-active) [64 Cu]Cu-pDefs are created with varying lipid acyl chain length and charge, representing the most prevalently studied microbubble compositions. In vitro, C16 chain length microbubbles yield 2-3x smaller nanoprogeny than C18 microbubbles post FUS. In vivo, [64 Cu]Cu-pDefs are tracked in healthy and 4T1 tumor-bearing mice ± FUS over 48 h qualitatively through fluorescence imaging (to characterize particle disruption) and quantitatively through PET and γ-counting. These studies reveal the impact of microbubble composition and FUS on microbubble dissolution rates, shell circulation, off-target tissue retention (predominantly the liver and spleen), and FUS enhancement of tumor delivery. These findings yield pharmacokinetic microbubble structure-activity relationships that disrupt conventional knowledge, the implications of which on MB-FUS platform design, safety, and nanomedicine delivery are discussed.
Collapse
Affiliation(s)
- Maneesha A. Rajora
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Institute of Biomedical EngineeringUniversity of TorontoTorontoOntarioM5G 1L7Canada
| | - Alexander Dhaliwal
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
| | - Mark Zheng
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
| | - Victor Choi
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
| | - Marta Overchuk
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Institute of Biomedical EngineeringUniversity of TorontoTorontoOntarioM5G 1L7Canada
- Joint Department of Biomedical EngineeringUniversity of North Carolina at Chapel Hill and North Carolina State UniversityChapel HillNC27599USA
| | - Jenny W. H. Lou
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
| | - Carly Pellow
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
- Sunnybrook Research InstituteTorontoOntarioM4N 3M5Canada
| | - David Goertz
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
- Sunnybrook Research InstituteTorontoOntarioM4N 3M5Canada
| | - Juan Chen
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
| | - Gang Zheng
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Institute of Biomedical EngineeringUniversity of TorontoTorontoOntarioM5G 1L7Canada
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
| |
Collapse
|
6
|
Porello I, Bono N, Candiani G, Cellesi F. Advancing nucleic acid delivery through cationic polymer design: non-cationic building blocks from the toolbox. Polym Chem 2024; 15:2800-2826. [DOI: 10.1039/d4py00234b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
The rational integration of non-cationic building blocks into cationic polymers can be devised to enhance the performance of the resulting gene delivery vectors, improving cell targeting behavior, uptake, endosomal escape, toxicity, and transfection efficiency.
Collapse
Affiliation(s)
- Ilaria Porello
- Department of Chemistry, Materials and Chemical Engineering “G. Natta”, Politecnico di Milano, Via Luigi Mancinelli 7, 20131, Milan, Italy
| | - Nina Bono
- Department of Chemistry, Materials and Chemical Engineering “G. Natta”, Politecnico di Milano, Via Luigi Mancinelli 7, 20131, Milan, Italy
| | - Gabriele Candiani
- Department of Chemistry, Materials and Chemical Engineering “G. Natta”, Politecnico di Milano, Via Luigi Mancinelli 7, 20131, Milan, Italy
| | - Francesco Cellesi
- Department of Chemistry, Materials and Chemical Engineering “G. Natta”, Politecnico di Milano, Via Luigi Mancinelli 7, 20131, Milan, Italy
| |
Collapse
|
7
|
Sylvers J, Wang Y, Yuan F. Nuclear Entry of DNA and Transgene Expression in Dividing and Non-dividing Cells. Cell Mol Bioeng 2023; 16:459-474. [PMID: 38099212 PMCID: PMC10716082 DOI: 10.1007/s12195-023-00784-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 09/03/2023] [Indexed: 12/17/2023] Open
Abstract
Introduction Plasmid DNA (pDNA) must be delivered into the nucleus for transgene expression in mammalian cells. The entry may happen passively during the nuclear envelope breakdown and reformation in dividing cells or actively through the nuclear pore complexes. The goal of this study was to investigate the relative importance of these two pathways for pDNA nuclear entry and subsequent gene expression. Methods To measure nuclear entry of pDNA encoding enhanced green florescence protein (EGFP) in electrotransfected cells, we developed a sensitive technique for quantitative analysis of pDNA in the nuclei, based on a hybridization probe for pDNA detection at the single molecule level and automatic image analysis. In matched experiments, we used an mRNA targeted hybridization probe to quantify reporter mRNA expression per cell, and flow cytometry to quantify expression of EGFP. Results We discovered two distinct patterns of pDNA distribution in the nuclei: punctate and diffuse, which were dominant in arrested and unarrested cells, respectively. The cell cycle arrest decreased diffuse pDNA and increased punctate pDNA. Its net effect was a decrease in the total intranuclear pDNA. Additionally, the cell cycle arrest increased the reporter mRNA synthesis but had no substantial impact on reporter protein expression. Conclusion Results from the study demonstrated that the efficient nuclear entry of pDNA during cell division did not necessarily lead to a high level of transgene expression. They also suggested that the punctate pDNA was more transcriptionally active than diffuse pDNA in the nuclei. These data will be useful in future studies for understanding mechanisms of nonviral gene delivery.
Collapse
Affiliation(s)
- Justin Sylvers
- Department of Biomedical Engineering, Duke University, Durham, NC 27708 USA
| | - Yifei Wang
- Department of Biomedical Engineering, Duke University, Durham, NC 27708 USA
| | - Fan Yuan
- Department of Biomedical Engineering, Duke University, Durham, NC 27708 USA
| |
Collapse
|
8
|
Loder S, Patel N, Morgani S, Sambon M, Leucht P, Levi B. Genetic models for lineage tracing in musculoskeletal development, injury, and healing. Bone 2023; 173:116777. [PMID: 37156345 PMCID: PMC10860167 DOI: 10.1016/j.bone.2023.116777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 04/07/2023] [Accepted: 04/17/2023] [Indexed: 05/10/2023]
Abstract
Musculoskeletal development and later post-natal homeostasis are highly dynamic processes, marked by rapid structural and functional changes across very short periods of time. Adult anatomy and physiology are derived from pre-existing cellular and biochemical states. Consequently, these early developmental states guide and predict the future of the system as a whole. Tools have been developed to mark, trace, and follow specific cells and their progeny either from one developmental state to the next or between circumstances of health and disease. There are now many such technologies alongside a library of molecular markers which may be utilized in conjunction to allow for precise development of unique cell 'lineages'. In this review, we first describe the development of the musculoskeletal system beginning as an embryonic germ layer and at each of the key developmental stages that follow. We then discuss these structures in the context of adult tissues during homeostasis, injury, and repair. Special focus is given in each of these sections to the key genes involved which may serve as markers of lineage or later in post-natal tissues. We then finish with a technical assessment of lineage tracing and the techniques and technologies currently used to mark cells, tissues, and structures within the musculoskeletal system.
Collapse
Affiliation(s)
- Shawn Loder
- Department of Plastic Surgery, University of Pittsburgh, Scaife Hall, Suite 6B, 3550 Terrace Street, Pittsburgh, PA 15261, USA
| | - Nicole Patel
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | | | | - Benjamin Levi
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
9
|
Kida H, Yamasaki Y, Feril Jr. LB, Endo H, Itaka K, Tachibana K. Efficient mRNA Delivery with Lyophilized Human Serum Albumin-Based Nanobubbles. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:1283. [PMID: 37049376 PMCID: PMC10097217 DOI: 10.3390/nano13071283] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/30/2023] [Accepted: 04/04/2023] [Indexed: 06/19/2023]
Abstract
In this study, we developed an efficient mRNA delivery vehicle by optimizing a lyophilization method for preserving human serum albumin-based nanobubbles (HSA-NBs), bypassing the need for artificial stabilizers. The morphology of the lyophilized material was verified using scanning electron microscopy, and the concentration, size, and mass of regenerated HSA-NBs were verified using flow cytometry, nanoparticle tracking analysis, and resonance mass measurements, and compared to those before lyophilization. The study also evaluated the response of HSA-NBs to 1 MHz ultrasound irradiation and their ultrasound (US) contrast effect. The functionality of the regenerated HSA-NBs was confirmed by an increased expression of intracellularly transferred Gluc mRNA, with increasing intensity of US irradiation. The results indicated that HSA-NBs retained their structural and functional integrity markedly, post-lyophilization. These findings support the potential of lyophilized HSA-NBs, as efficient imaging, and drug delivery systems for various medical applications.
Collapse
Affiliation(s)
- Hiroshi Kida
- Department of Anatomy, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Yutaro Yamasaki
- Department of Anatomy, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Loreto B. Feril Jr.
- Department of Anatomy, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Hitomi Endo
- Department of Anatomy, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Keiji Itaka
- Department of Biofunction Research, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Tokyo 101-0062, Japan
| | - Katsuro Tachibana
- Department of Anatomy, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| |
Collapse
|
10
|
Opportunities and challenges in delivering biologics for Alzheimer's disease by low-intensity ultrasound. Adv Drug Deliv Rev 2022; 189:114517. [PMID: 36030018 DOI: 10.1016/j.addr.2022.114517] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/15/2022] [Accepted: 08/19/2022] [Indexed: 01/24/2023]
Abstract
Low-intensity ultrasound combined with intravenously injected microbubbles (US+MB) is a novel treatment modality for brain disorders, including Alzheimer's disease (AD), safely and transiently allowing therapeutic agents to overcome the blood-brain barrier (BBB) that constitutes a major barrier for therapeutic agents. Here, we first provide an update on immunotherapies in AD and how US+MB has been applied to AD mouse models and in clinical trials, considering the ultrasound and microbubble parameter space. In the second half of the review, we compare different in vitro BBB models and discuss strategies for combining US+MB with BBB modulators (targeting molecules such as claudin-5), and highlight the insight provided by super-resolution microscopy. Finally, we conclude with a short discussion on how in vitro findings can inform the design of animal studies, and how the insight gained may aid treatment optimization in the clinical ultrasound space.
Collapse
|
11
|
Germain P, Delalande A, Pichon C. Role of Muscle LIM Protein in Mechanotransduction Process. Int J Mol Sci 2022; 23:ijms23179785. [PMID: 36077180 PMCID: PMC9456170 DOI: 10.3390/ijms23179785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/14/2022] [Accepted: 08/26/2022] [Indexed: 11/25/2022] Open
Abstract
The induction of protein synthesis is crucial to counteract the deconditioning of neuromuscular system and its atrophy. In the past, hormones and cytokines acting as growth factors involved in the intracellular events of these processes have been identified, while the implications of signaling pathways associated with the anabolism/catabolism ratio in reference to the molecular mechanism of skeletal muscle hypertrophy have been recently identified. Among them, the mechanotransduction resulting from a mechanical stress applied to the cell appears increasingly interesting as a potential pathway for therapeutic intervention. At present, there is an open question regarding the type of stress to apply in order to induce anabolic events or the type of mechanical strain with respect to the possible mechanosensing and mechanotransduction processes involved in muscle cells protein synthesis. This review is focused on the muscle LIM protein (MLP), a structural and mechanosensing protein with a LIM domain, which is expressed in the sarcomere and costamere of striated muscle cells. It acts as a transcriptional cofactor during cell proliferation after its nuclear translocation during the anabolic process of differentiation and rebuilding. Moreover, we discuss the possible opportunity of stimulating this mechanotransduction process to counteract the muscle atrophy induced by anabolic versus catabolic disorders coming from the environment, aging or myopathies.
Collapse
Affiliation(s)
- Philippe Germain
- UFR Sciences and Techniques, University of Orleans, 45067 Orleans, France
- Center for Molecular Biophysics, CNRS Orleans, 45071 Orleans, France
| | - Anthony Delalande
- UFR Sciences and Techniques, University of Orleans, 45067 Orleans, France
- Center for Molecular Biophysics, CNRS Orleans, 45071 Orleans, France
| | - Chantal Pichon
- UFR Sciences and Techniques, University of Orleans, 45067 Orleans, France
- Center for Molecular Biophysics, CNRS Orleans, 45071 Orleans, France
- Institut Universitaire de France, 1 Rue Descartes, 75231 Paris, France
- Correspondence:
| |
Collapse
|
12
|
Du M, Li Y, Chen Z. Sonoporation-Mediated Gene Transfection: A Novel Direction for Cell Reprogramming In Vivo. Front Bioeng Biotechnol 2022; 9:803055. [PMID: 35174147 PMCID: PMC8841490 DOI: 10.3389/fbioe.2021.803055] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/09/2021] [Indexed: 11/27/2022] Open
Affiliation(s)
- Meng Du
- The First Affiliated Hospital, Medical Imaging Centre, Hengyang Medical School, University of South China, Hengyang, China
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China
| | - Yue Li
- The First Affiliated Hospital, Medical Imaging Centre, Hengyang Medical School, University of South China, Hengyang, China
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China
- Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhiyi Chen
- The First Affiliated Hospital, Medical Imaging Centre, Hengyang Medical School, University of South China, Hengyang, China
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China
- *Correspondence: Zhiyi Chen,
| |
Collapse
|
13
|
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
|
14
|
Rapid Magneto-Sonoporation of Adipose-Derived Cells. MATERIALS 2021; 14:ma14174877. [PMID: 34500968 PMCID: PMC8432646 DOI: 10.3390/ma14174877] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/23/2021] [Accepted: 08/24/2021] [Indexed: 01/03/2023]
Abstract
By permeabilizing the cell membrane with ultrasound and facilitating the uptake of iron oxide nanoparticles, the magneto-sonoporation (MSP) technique can be used to instantaneously label transplantable cells (like stem cells) to be visualized via magnetic resonance imaging in vivo. However, the effects of MSP on cells are still largely unexplored. Here, we applied MSP to the widely applicable adipose-derived stem cells (ASCs) for the first time and investigated its effects on the biology of those cells. Upon optimization, MSP allowed us to achieve a consistent nanoparticle uptake (in the range of 10 pg/cell) and a complete membrane resealing in few minutes. Surprisingly, this treatment altered the metabolic activity of cells and induced their differentiation towards an osteoblastic profile, as demonstrated by an increased expression of osteogenic genes and morphological changes. Histological evidence of osteogenic tissue development was collected also in 3D hydrogel constructs. These results point to a novel role of MSP in remote biophysical stimulation of cells with focus application in bone tissue repair.
Collapse
|
15
|
Liufu C, Li Y, Lin Y, Yu J, Du M, Chen Y, Yang Y, Gong X, Chen Z. Synergistic ultrasonic biophysical effect-responsive nanoparticles for enhanced gene delivery to ovarian cancer stem cells. Drug Deliv 2021; 27:1018-1033. [PMID: 32627597 PMCID: PMC8216435 DOI: 10.1080/10717544.2020.1785583] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Ovarian cancer stem cells (OCSCs) that are a subpopulation within bulk tumor survive chemotherapy and conduce to chemo-resistance and tumor relapse. However, conventional gene delivery is unsuitable for the on-demand content release, which limits OCSCs therapeutic utility. Here, we reported ultrasound-targeted microbubble destruction (UTMD)-triggerable poly(ethylene glycol)-disulfide bond-polyethylenimine loaded microbubble (PSP@MB). Taking advantage of glutathione (GSH) responsiveness, ultrasound triggering and spatiotemporally controlled release manner, PSP@MB is expected to realize local gene delivery for OCSCs treatment. But the biophysical mechanisms of gene delivery via PSP@MB and ultrasound remain unknown. The aim of this study is to determine the potential of gene delivery to OCSCs via ultrasonic synergistic biophysical effects and GSH-sensitive PSP@MB. The GSH-sensitive disulfide bond cleavable properties of PSP@MB were confirmed by 1H NMR spectra and infrared spectroscopy. The biophysical mechanisms between PSP@MB and cells were confirmed by scanning electron microscopy (SEM) and confocal laser scanning microscope (CLSM) to optimize the ultrasonic gene delivery system. The gene transfection via ultrasound and PSP@MB was closely related to the biophysical mechanisms (sonoporation, enhanced-endocytosis, sonoprinting, and endosomal escape). Ultrasound combined with PSP@MB successfully delivered aldehyde dehydrogenase 1 (ALDH1) short hairpin RNA (shRNA) plasmid to OCSCs and promoted apoptosis of OCSCs. The gene transfection rate and apoptosis rate were (18.41 ± 2.41)% and (32.62 ± 2.36)% analyzed by flow cytometry separately. This study showed that ultrasound triggering and GSH responsive PSP@MB might provide a novel strategy for OCSCs treatment via sonoporation and enhanced-endocytosis.
Collapse
Affiliation(s)
- Chun Liufu
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, The Liwan Hospital of the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yue Li
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, The Liwan Hospital of the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yan Lin
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, The Liwan Hospital of the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jinsui Yu
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, The Liwan Hospital of the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Meng Du
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, The Liwan Hospital of the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuhao Chen
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, The Liwan Hospital of the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yaozhang Yang
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, The Liwan Hospital of the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaojing Gong
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen University Town, Shenzhen, China
| | - Zhiyi Chen
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, The Liwan Hospital of the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
16
|
Gailliègue FN, Tamošiūnas M, André FM, Mir LM. A Setup for Microscopic Studies of Ultrasounds Effects on Microliters Scale Samples: Analytical, Numerical and Experimental Characterization. Pharmaceutics 2021; 13:pharmaceutics13060847. [PMID: 34201070 PMCID: PMC8227135 DOI: 10.3390/pharmaceutics13060847] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 11/23/2022] Open
Abstract
Sonoporation is the process of cell membrane permeabilization, due to exposure to ultrasounds. There is a lack of consensus concerning the mechanisms of sonoporation: Understanding the mechanisms of sonoporation refines the choice of the ultrasonic parameters to be applied on the cells. Cells’ classical exposure systems to ultrasounds have several drawbacks, like the immersion of the cells in large volumes of liquid, the nonhomogeneous acoustic pressure in the large sample, and thus, the necessity for magnetic stirring to somehow homogenize the exposure of the cells. This article reports the development and characterization of a novel system allowing the exposure to ultrasounds of very small volumes and their observation under the microscope. The observation under a microscope imposes the exposure of cells and Giant Unilamellar Vesicles under an oblique incidence, as well as the very unusual presence of rigid walls limiting the sonicated volume. The advantages of this new setup are not only the use of a very small volume of cells culture medium/microbubbles (MB), but the presence of flat walls near the sonicated region that results in a more homogeneous ultrasonic pressure field, and thus, the control of the focal distance and the real exposure time. The setup presented here comprises the ability to survey the geometrical and dynamical aspects of the exposure of cells and MB to ultrasounds, if an ultrafast camera is used. Indeed, the setup thus fulfills all the requirements to apply ultrasounds conveniently, for accurate mechanistic experiments under an inverted fluorescence microscope, and it could have interesting applications in photoacoustic research.
Collapse
Affiliation(s)
- Florian N. Gailliègue
- Institut Gustave Roussy, Metabolic and Systemic Aspects of the Oncogenesis (METSY), Université Paris-Saclay, CNRS, 94805 Villejuif, France; (F.N.G.); (F.M.A.)
| | - Mindaugas Tamošiūnas
- Biophotonics Laboratory, Institute of Atomic Physics and Spectroscopy, University of Latvia, 19 Raina Blvd., LV-1586 Rīga, Latvia;
| | - Franck M. André
- Institut Gustave Roussy, Metabolic and Systemic Aspects of the Oncogenesis (METSY), Université Paris-Saclay, CNRS, 94805 Villejuif, France; (F.N.G.); (F.M.A.)
| | - Lluis M. Mir
- Institut Gustave Roussy, Metabolic and Systemic Aspects of the Oncogenesis (METSY), Université Paris-Saclay, CNRS, 94805 Villejuif, France; (F.N.G.); (F.M.A.)
- Correspondence: ; Tel.: +33-(0)1421-14792
| |
Collapse
|
17
|
Li Y, Chen Z, Ge S. Sonoporation: Underlying Mechanisms and Applications in Cellular Regulation. BIO INTEGRATION 2021. [DOI: 10.15212/bioi-2020-0028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Ultrasound combined with microbubble-mediated sonoporation has been applied to enhance drug or gene intracellular delivery. Sonoporation leads to the formation of openings in the cell membrane, triggered by ultrasound-mediated oscillations and destruction of microbubbles. Multiple mechanisms
are involved in the occurrence of sonoporation, including ultrasonic parameters, microbubbles size, and the distance of microbubbles to cells. Recent advances are beginning to extend applications through the assistance of contrast agents, which allow ultrasound to connect directly to cellular
functions such as gene expression, cellular apoptosis, differentiation, and even epigenetic reprogramming. In this review, we summarize the current state of the art concerning microbubble‐cell interactions and sonoporation effects leading to cellular functions.
Collapse
Affiliation(s)
- Yue Li
- First Affiliated Hospital of University of South China, Hengyang, China
| | - Zhiyi Chen
- First Affiliated Hospital of University of South China, Hengyang, China
| | - Shuping Ge
- Department of Pediatrics, St Christopher’s Hospital for Children, Tower Health and Drexel University, Philadelphia, PA (S.G.)
| |
Collapse
|
18
|
Dwivedi P, Kiran S, Han S, Dwivedi M, Khatik R, Fan R, Mangrio FA, Du K, Zhu Z, Yang C, Huang F, Ejaz A, Han R, Si T, Xu RX. Magnetic Targeting and Ultrasound Activation of Liposome-Microbubble Conjugate for Enhanced Delivery of Anticancer Therapies. ACS APPLIED MATERIALS & INTERFACES 2020; 12:23737-23751. [PMID: 32374147 DOI: 10.1021/acsami.0c05308] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Effective delivery of chemotherapeutics with minimal toxicity and maximal outcome is clinically important but technically challenging. Here, we synthesize a complex of doxorubicin (DOX)-loaded magneto-liposome (DOX-ML) microbubbles (DOX-ML-MBs) for magnetically responsive and ultrasonically sensitive delivery of anticancer therapies with enhanced efficiency. Citrate-stabilized iron oxide nanoparticles (MNs) of 6.8 ± 1.36 nm were synthesized, loaded with DOX in the core of oligolamellar vesicles of 172 ± 9.2 nm, and covalently conjugated with perfluorocarbon (PFC)-gas-loaded microbubbles to form DOX-ML-MBs of ∼4 μm. DOX-ML-MBs exhibited significant magnetism and were able to release chemotherapeutics and DOX-MLs instantly upon exposure to ultrasound (US) pulses. In vitro studies showed that DOX-ML-MBs in the presence of US pulses promoted apoptosis and were highly effective in killing both BxPc-3 and Panc02 pancreatic cancer cells even at a low dose. Significant reduction in the tumor volume was observed after intravenous administration of DOX-ML-MBs in comparison to the control group in a pancreatic cancer xenograft model of nude mice. Deeply penetrated iron oxide nanoparticles throughout the magnetically targeted tumor tissues in the presence of US stimulation were clearly observed. Our study demonstrated the potential of using DOX-ML-MBs for site-specific targeting and controlled drug release. It opens a new avenue for the treatment of pancreatic cancer and other tissue malignancies where precise delivery of therapeutics is necessary.
Collapse
Affiliation(s)
- Pankaj Dwivedi
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
- Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
| | - Sonia Kiran
- Department of Surgery, The Ohio State University, Columbus, Ohio 43210, United States
| | - Shuya Han
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
- Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
| | - Monika Dwivedi
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
- Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
| | - Renuka Khatik
- Hefei National Laboratory of Physical Sciences at Microscale, Department of Chemistry, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, P. R. China
| | - Rong Fan
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
- Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
| | - Farhana Akbar Mangrio
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
- Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
| | - Kun Du
- Department of Electronic Science and Technology, University of Science and Technology of China Hefei, Hefei, Anhui 230027, P. R. China
| | - Zhiqiang Zhu
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
- Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
| | - Chaoyu Yang
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
- Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
| | - Fangsheng Huang
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
- Hefei National Laboratory of Physical Sciences at Microscale, Department of Chemistry, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, P. R. China
| | - Aslam Ejaz
- Department of Surgery, The Ohio State University, Columbus, Ohio 43210, United States
| | - Renzhi Han
- Department of Surgery, The Ohio State University, Columbus, Ohio 43210, United States
| | - Ting Si
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
| | - Ronald X Xu
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
19
|
Belling JN, Heidenreich LK, Tian Z, Mendoza AM, Chiou TT, Gong Y, Chen NY, Young TD, Wattanatorn N, Park JH, Scarabelli L, Chiang N, Takahashi J, Young SG, Stieg AZ, De Oliveira S, Huang TJ, Weiss PS, Jonas SJ. Acoustofluidic sonoporation for gene delivery to human hematopoietic stem and progenitor cells. Proc Natl Acad Sci U S A 2020; 117:10976-10982. [PMID: 32358194 PMCID: PMC7245081 DOI: 10.1073/pnas.1917125117] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Advances in gene editing are leading to new medical interventions where patients' own cells are used for stem cell therapies and immunotherapies. One of the key limitations to translating these treatments to the clinic is the need for scalable technologies for engineering cells efficiently and safely. Toward this goal, microfluidic strategies to induce membrane pores and permeability have emerged as promising techniques to deliver biomolecular cargo into cells. As these technologies continue to mature, there is a need to achieve efficient, safe, nontoxic, fast, and economical processing of clinically relevant cell types. We demonstrate an acoustofluidic sonoporation method to deliver plasmids to immortalized and primary human cell types, based on pore formation and permeabilization of cell membranes with acoustic waves. This acoustofluidic-mediated approach achieves fast and efficient intracellular delivery of an enhanced green fluorescent protein-expressing plasmid to cells at a scalable throughput of 200,000 cells/min in a single channel. Analyses of intracellular delivery and nuclear membrane rupture revealed mechanisms underlying acoustofluidic delivery and successful gene expression. Our studies show that acoustofluidic technologies are promising platforms for gene delivery and a useful tool for investigating membrane repair.
Collapse
Affiliation(s)
- Jason N Belling
- California NanoSystems Institute, University of California, Los Angeles, CA 90095
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095
| | - Liv K Heidenreich
- California NanoSystems Institute, University of California, Los Angeles, CA 90095
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095
| | - Zhenhua Tian
- Department of Mechanical Engineering and Material Science, Duke University, Durham, NC 27707
- Department of Aerospace Engineering, Mississippi State University, Starkville, MS 39762
| | - Alexandra M Mendoza
- California NanoSystems Institute, University of California, Los Angeles, CA 90095
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095
| | - Tzu-Ting Chiou
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
- Children's Discovery and Innovation Institute, University of California, Los Angeles, CA 90095
| | - Yao Gong
- California NanoSystems Institute, University of California, Los Angeles, CA 90095
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095
| | - Natalie Y Chen
- Department of Medicine and the Molecular Biology Institute, University of California, Los Angeles, CA 90095
- Department of Human Genetics and the Molecular Biology Institute, University of California, Los Angeles, CA 90095
| | - Thomas D Young
- California NanoSystems Institute, University of California, Los Angeles, CA 90095
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095
| | - Natcha Wattanatorn
- California NanoSystems Institute, University of California, Los Angeles, CA 90095
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095
| | - Jae Hyeon Park
- California NanoSystems Institute, University of California, Los Angeles, CA 90095
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095
| | - Leonardo Scarabelli
- California NanoSystems Institute, University of California, Los Angeles, CA 90095
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095
| | - Naihao Chiang
- California NanoSystems Institute, University of California, Los Angeles, CA 90095
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095
| | - Jack Takahashi
- California NanoSystems Institute, University of California, Los Angeles, CA 90095
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095
| | - Stephen G Young
- Department of Medicine and the Molecular Biology Institute, University of California, Los Angeles, CA 90095
| | - Adam Z Stieg
- California NanoSystems Institute, University of California, Los Angeles, CA 90095
| | - Satiro De Oliveira
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
- Children's Discovery and Innovation Institute, University of California, Los Angeles, CA 90095
| | - Tony Jun Huang
- Department of Mechanical Engineering and Material Science, Duke University, Durham, NC 27707
| | - Paul S Weiss
- California NanoSystems Institute, University of California, Los Angeles, CA 90095;
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095
- Department of Materials Science and Engineering, University of California, Los Angeles, CA 90095
| | - Steven J Jonas
- California NanoSystems Institute, University of California, Los Angeles, CA 90095;
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
- Children's Discovery and Innovation Institute, University of California, Los Angeles, CA 90095
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA 90095
| |
Collapse
|
20
|
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
|
21
|
Mignet N, Marie C, Delalande A, Manta S, Bureau MF, Renault G, Scherman D, Pichon C. Microbubbles for Nucleic Acid Delivery in Liver Using Mild Sonoporation. Methods Mol Biol 2019; 1943:377-387. [PMID: 30838630 DOI: 10.1007/978-1-4939-9092-4_25] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Ultrasound-mediated gene delivery is an interesting approach, which could help in increasing gene transfer in deep tissues. Moreover, it allows for performing experiments guided by the image to determine which elements are required. Microbubbles complexed with a eukaryotic expression cassette are excellent agents as they are responsive to ultrasounds and, upon oscillation, can destabilize membranes to enhance gene transfer. Here, we describe the preparation of positively charged microbubbles, plasmid free of antibiotic resistance marker, their combination and the conditions of ultrasound-mediated liver transfection post-systemic administration in mice. This association allowed us to obtain a superior liver gene expression at least over 8 months after a single injection.
Collapse
Affiliation(s)
- Nathalie Mignet
- Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), INSERM, U1022, Paris, France. .,CNRS, UMR8258, Paris, France. .,Faculté de Pharmacie, Sorbonne Paris Cité, Université Paris Descartes, Paris, France. .,Chimie ParisTech, PSL Research University, Paris, France.
| | - Corinne Marie
- INSERM, U1022, Paris, France.,CNRS, UMR8258, Paris, France.,Faculté de Pharmacie, Sorbonne Paris Cité, Université Paris Descartes, Paris, France.,Chimie ParisTech, PSL Research University, Paris, France
| | - Anthony Delalande
- Centre de Biophysique Moléculaire and Université d'Orléans, CNRS-UPR 4301, Orléans, France
| | - Simona Manta
- INSERM, U1022, Paris, France.,CNRS, UMR8258, Paris, France.,Faculté de Pharmacie, Sorbonne Paris Cité, Université Paris Descartes, Paris, France.,Chimie ParisTech, PSL Research University, Paris, France
| | - Michel-Francis Bureau
- INSERM, U1022, Paris, France.,CNRS, UMR8258, Paris, France.,Faculté de Pharmacie, Sorbonne Paris Cité, Université Paris Descartes, Paris, France.,Chimie ParisTech, PSL Research University, Paris, France
| | - Gilles Renault
- INSERM, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France.,Sorbonne Paris Cité, Université Paris Descartes, Paris, France
| | - Daniel Scherman
- INSERM, U1022, Paris, France.,CNRS, UMR8258, Paris, France.,Faculté de Pharmacie, Sorbonne Paris Cité, Université Paris Descartes, Paris, France.,Chimie ParisTech, PSL Research University, Paris, France
| | - Chantal Pichon
- Centre de Biophysique Moléculaire and Université d'Orléans, CNRS-UPR 4301, Orléans, France
| |
Collapse
|
22
|
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
|
23
|
D'Arrigo JS. Targeting Early Dementia: Using Lipid Cubic Phase Nanocarriers to Cross the Blood⁻Brain Barrier. Biomimetics (Basel) 2018; 3:E4. [PMID: 31105226 PMCID: PMC6352688 DOI: 10.3390/biomimetics3010004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 02/21/2018] [Accepted: 03/06/2018] [Indexed: 12/14/2022] Open
Abstract
Over the past decades, a frequent co-morbidity of cerebrovascular pathology and Alzheimer's disease has been observed. Numerous published studies indicate that the preservation of a healthy cerebrovascular endothelium can be an important therapeutic target. By incorporating the appropriate drug(s) into biomimetic (lipid cubic phase) nanocarriers, one obtains a multitasking combination therapeutic, which targets certain cell surface scavenger receptors, mainly class B type I (i.e., SR-BI), and crosses the blood⁻brain barrier. This targeting allows for various cell types related to Alzheimer's to be simultaneously searched out for localized drug treatment in vivo.
Collapse
|
24
|
Boehringer S, Ruzgys P, Tamò L, Šatkauskas S, Geiser T, Gazdhar A, Hradetzky D. A new electrospray method for targeted gene delivery. Sci Rep 2018; 8:4031. [PMID: 29507307 PMCID: PMC5838090 DOI: 10.1038/s41598-018-22280-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 02/20/2018] [Indexed: 11/15/2022] Open
Abstract
A challenge for gene therapy is absence of safe and efficient local delivery of therapeutic genetic material. An efficient and reproducible physical method of electrospray for localized and targeted gene delivery is presented. Electrospray works on the principle of coulombs repulsion, under influence of electric field the liquid carrying genetic material is dispersed into micro droplets and is accelerated towards the targeted tissue, acting as a counter electrode. The accelerated droplets penetrate the targeted cells thus facilitating the transfer of genetic material into the cell. The work described here presents the principle of electrospray for gene delivery, the basic instrument design, and the various optimized parameters to enhance gene transfer in vitro. We estimate a transfection efficiency of up to 60% was achieved. We describe an efficient gene transfer method and a potential electrospray-mediated gene transfer mechanism.
Collapse
Affiliation(s)
- Stephan Boehringer
- Institute for Medical and Analytical Technologies, School of Life Sciences, University of Applied Sciences and Arts Northwestern Switzerland, Muttenz, Switzerland
| | - Paulius Ruzgys
- Department of Pulmonary Medicine, University Hospital Bern, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Bern, Switzerland
- Biophysical Research Group, Faculty of Natural Sciences, Vytautas Magnus University, Kaunas, Lithuania
| | - Luca Tamò
- Department of Pulmonary Medicine, University Hospital Bern, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Science, University of Bern, Bern, Switzerland
| | - Saulius Šatkauskas
- Biophysical Research Group, Faculty of Natural Sciences, Vytautas Magnus University, Kaunas, Lithuania
| | - Thomas Geiser
- Department of Pulmonary Medicine, University Hospital Bern, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Amiq Gazdhar
- Department of Pulmonary Medicine, University Hospital Bern, Bern, Switzerland.
- Department of Biomedical Research, University of Bern, Bern, Switzerland.
| | - David Hradetzky
- Institute for Medical and Analytical Technologies, School of Life Sciences, University of Applied Sciences and Arts Northwestern Switzerland, Muttenz, Switzerland.
| |
Collapse
|
25
|
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
|
26
|
Nanotherapy for Alzheimer's disease and vascular dementia: Targeting senile endothelium. Adv Colloid Interface Sci 2018; 251:44-54. [PMID: 29274774 DOI: 10.1016/j.cis.2017.12.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 12/01/2017] [Accepted: 12/02/2017] [Indexed: 12/12/2022]
Abstract
Due to the complexity of Alzheimer's disease, multiple cellular types need to be targeted simultaneously in order for a given therapy to demonstrate any major effectiveness. Ultrasound-sensitive coated microbubbles (in a targeted lipid nanoemulsion) are available. Versatile small molecule drug(s) targeting multiple pathways of Alzheimer's disease pathogenesis are known. By incorporating such drug(s) into the targeted "lipid-coated microbubble" [LCM]/"nanoparticle-derived" [ND] (or LCM/ND) nanoemulsion type, one obtains a multitasking combination therapeutic for translational medicine. This multitasking therapeutic targets cell-surface scavenger receptors (mainly class B type I), or SR-BI, making possible for various Alzheimer's-related cell types to be simultaneously searched out for localized drug treatment in vivo. Besides targeting cell-surface SR-BI, the proposed LCM/ND-nanoemulsion combination therapeutic(s) include a characteristic lipid-coated microbubble [LCM] subpopulation (i.e., a stable LCM suspension); such film-stabilized microbubbles are well known to substantially reduce the acoustic power levels needed for accomplishing temporary noninvasive (transcranial) ultrasound treatment, or sonoporation, if additionally desired for the Alzheimer's patient.
Collapse
|
27
|
Cationic gas-filled microbubbles for ultrasound-based nucleic acids delivery. Biosci Rep 2017; 37:BSR20160619. [PMID: 29180378 PMCID: PMC5741830 DOI: 10.1042/bsr20160619] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 11/23/2017] [Accepted: 11/27/2017] [Indexed: 12/11/2022] Open
Abstract
The use of ultrasound has gained great interest for nucleic acids delivery. Ultrasound can reach deep tissues in non-invasive manner. The process of sonoporation is based on the use of low-frequency ultrasound combined with gas-filled microbubbles (MBs) allowing an improved delivery of molecules including nucleic acids in the insonified tissue. For in vivo gene transfer, the engineering of cationic MBs is essential for creating strong electrostatic interactions between MBs and nucleic acids leading to their protection against nucleases degradation and high concentration within the target tissue. Cationic MBs must be stable enough to withstand nucleic acids interaction, have a good size distribution for in vivo administration, and enough acoustic activity to be detected by echography. This review aims to summarize the basic principles of ultrasound-based delivery and new knowledge acquired in these recent years about this method. A focus is made on gene delivery by discussing reported studies made with cationic MBs including ours. They have the ability for efficient delivery of plasmid DNA (pDNA), mRNA or siRNA. Last, we discuss about the key challenges that have to be faced for a fine use of this delivery system.
Collapse
|
28
|
Alzheimer’s Disease, Brain Injury, and C.N.S. Nanotherapy in Humans: Sonoporation Augmenting Drug Targeting. Med Sci (Basel) 2017. [PMCID: PMC5753658 DOI: 10.3390/medsci5040029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Owing to the complexity of neurodegenerative diseases, multiple cellular types need to be targeted simultaneously in order for a given therapy to demonstrate any major effectiveness. Ultrasound-sensitive coated microbubbles (in a targeted nanoemulsion) are available. Versatile small-molecule drug(s) targeting multiple pathways of Alzheimer’s disease pathogenesis are known. By incorporating such drug(s) into the targeted lipid-coated microbubble/nanoparticle-derived (LCM/ND) lipid nanoemulsion type, one obtains a multitasking combination therapeutic for translational medicine. This multitasking therapeutic targets cell-surface scavenger receptors (mainly scavenger receptor class B type I (SR-BI)), making it possible for various Alzheimer’s-related cell types to be simultaneously sought for localized drug treatment in vivo. Besides targeting cell-surface SR-BI, the proposed LCM/ND-nanoemulsion combination therapeutic(s) include a characteristic lipid-coated microbubble (LCM) subpopulation (i.e., a stable LCM suspension); such LCM substantially reduce the acoustic power levels needed for accomplishing temporary noninvasive (transcranial) ultrasound treatment, or sonoporation, if additionally desired for the Alzheimer’s patient.
Collapse
|
29
|
Enhancing the cytotoxicity of chemoradiation with radiation-guided delivery of anti-MGMT morpholino oligonucleotides in non-methylated solid tumors. Cancer Gene Ther 2017; 24:348-357. [PMID: 28752860 PMCID: PMC5605678 DOI: 10.1038/cgt.2017.27] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 05/20/2017] [Indexed: 01/25/2023]
Abstract
The DNA repair enzyme O6-methylguanine DNA methyltransferase (MGMT) is epigenetically silenced in some tumors by MGMT gene promoter methylation. MGMT-hypermethylated solid tumors have enhanced susceptibility to the cytotoxic effects of alkylating chemotherapy such as temozolomide, compared with non-methylated tumors. In glioblastoma, subjects with MGMT hypermethylation have significantly longer survival rates after chemoradiotherapy. We report the first successful use of a non-ablative dose of ionizing radiation to prime human cancer cells to enhance the uptake of unmodified anti-MGMT morpholino oligonucleotide (AMON) sequences. We demonstrate >40% reduction in the in vitro proliferation index and cell viability in radiation-primed MGMT-expressing human solid tumor cells treated with a single dose of AMONs and temozolomide. We further demonstrate the feasibility of using a non-ablative dose of radiation in vivo to guide and enhance the delivery of intravenously administered AMONs to achieve 50% MGMT knockdown only at radiation-primed tumor sites in a subcutaneous tumor model. Local upregulation of physiological endocytosis after radiation may have a role in radiation-guided uptake of AMONs. This approach holds direct translational significance in glioblastoma and brain metastases where radiation is part of the standard of care; our approach to silence MGMT could overcome the significant problem of MGMT-mediated chemoresistance.
Collapse
|
30
|
Manta S, Renault G, Delalande A, Couture O, Lagoutte I, Seguin J, Lager F, Houzé P, Midoux P, Bessodes M, Scherman D, Bureau MF, Marie C, Pichon C, Mignet N. Cationic microbubbles and antibiotic-free miniplasmid for sustained ultrasound-mediated transgene expression in liver. J Control Release 2017; 262:170-181. [PMID: 28710005 DOI: 10.1016/j.jconrel.2017.07.015] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 07/06/2017] [Accepted: 07/09/2017] [Indexed: 11/15/2022]
Abstract
Despite the increasing number of clinical trials in gene therapy, no ideal methods still allow non-viral gene transfer in deep tissues such as the liver. We were interested in ultrasound (US)-mediated gene delivery to provide long term liver expression. For this purpose, new positively charged microbubbles were designed and complexed with pFAR4, a highly efficient small length miniplasmid DNA devoid of antibiotic resistance sequence. Sonoporation parameters, such as insonation time, acoustic pressure and duration of plasmid injection were controlled under ultrasound imaging guidance. The optimization of these various parameters was performed by bioluminescence optical imaging of luciferase reporter gene expression in the liver. Mice were injected with 50μg pFAR4-LUC either alone, or complexed with positively charged microbubbles, or co-injected with neutral MicroMarker™ microbubbles, followed by low ultrasound energy application to the liver. Injection of the pFAR4 encoding luciferase alone led to a transient transgene expression that lasted only for two days. The significant luciferase signal obtained with neutral microbubbles decreased over 2days and reached a plateau with a level around 1 log above the signal obtained with pFAR4 alone. With the newly designed positively charged microbubbles, we obtained a much stronger bioluminescence signal which increased over 2days. The 12-fold difference (p<0.05) between MicroMarker™ and our positively charged microbubbles was maintained over a period of 6months. Noteworthy, the positively charged microbubbles led to an improvement of 180-fold (p<0.001) as regard to free pDNA using unfocused ultrasound performed at clinically tolerated ultrasound amplitude. Transient liver damage was observed when using the cationic microbubble-pFAR4 complexes and the optimized sonoporation parameters. Immunohistochemistry analyses were performed to determine the nature of cells transfected. The pFAR4 miniplasmid complexed with cationic microbubbles allowed to transfect mostly hepatocytes compared to its co-injection with MicroMarker™ which transfected more preferentially endothelial cells.
Collapse
Affiliation(s)
- Simona Manta
- CNRS, UTCBS UMR 8258, F-75006 Paris, France; Université Paris Descartes, Sorbonne-Paris-Cité, UTCBS, F-75006 Paris, France; Chimie ParisTech, PSL Research University, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), F-75005 Paris, France; INSERM, UTCBS U 1022, F-75006 Paris, France
| | - Gilles Renault
- INSERM, U1016, Institut Cochin, Paris, France; CNRS, UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, France
| | - Anthony Delalande
- Centre de Biophysique Moléculaire and Université d'Orléans, UPR 4301, F-45071 Orléans, France
| | - Olivier Couture
- Institut Langevin - Ondes et Images, ESPCI ParisTech, PSL Research University, CNRS UMR7587, INSERM U979, 1, rue Jussieu, 75238 Paris, Cedex 05, France
| | - Isabelle Lagoutte
- INSERM, U1016, Institut Cochin, Paris, France; CNRS, UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, France
| | - Johanne Seguin
- CNRS, UTCBS UMR 8258, F-75006 Paris, France; Université Paris Descartes, Sorbonne-Paris-Cité, UTCBS, F-75006 Paris, France; Chimie ParisTech, PSL Research University, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), F-75005 Paris, France; INSERM, UTCBS U 1022, F-75006 Paris, France
| | - Franck Lager
- INSERM, U1016, Institut Cochin, Paris, France; CNRS, UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, France
| | - Pascal Houzé
- CNRS, UTCBS UMR 8258, F-75006 Paris, France; Université Paris Descartes, Sorbonne-Paris-Cité, UTCBS, F-75006 Paris, France; Chimie ParisTech, PSL Research University, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), F-75005 Paris, France; INSERM, UTCBS U 1022, F-75006 Paris, France
| | - Patrick Midoux
- Centre de Biophysique Moléculaire and Université d'Orléans, UPR 4301, F-45071 Orléans, France
| | - Michel Bessodes
- CNRS, UTCBS UMR 8258, F-75006 Paris, France; Université Paris Descartes, Sorbonne-Paris-Cité, UTCBS, F-75006 Paris, France; Chimie ParisTech, PSL Research University, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), F-75005 Paris, France; INSERM, UTCBS U 1022, F-75006 Paris, France
| | - Daniel Scherman
- CNRS, UTCBS UMR 8258, F-75006 Paris, France; Université Paris Descartes, Sorbonne-Paris-Cité, UTCBS, F-75006 Paris, France; Chimie ParisTech, PSL Research University, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), F-75005 Paris, France; INSERM, UTCBS U 1022, F-75006 Paris, France
| | - Michel-Francis Bureau
- CNRS, UTCBS UMR 8258, F-75006 Paris, France; Université Paris Descartes, Sorbonne-Paris-Cité, UTCBS, F-75006 Paris, France; Chimie ParisTech, PSL Research University, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), F-75005 Paris, France; INSERM, UTCBS U 1022, F-75006 Paris, France
| | - Corinne Marie
- CNRS, UTCBS UMR 8258, F-75006 Paris, France; Université Paris Descartes, Sorbonne-Paris-Cité, UTCBS, F-75006 Paris, France; Chimie ParisTech, PSL Research University, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), F-75005 Paris, France; INSERM, UTCBS U 1022, F-75006 Paris, France
| | - Chantal Pichon
- Centre de Biophysique Moléculaire and Université d'Orléans, UPR 4301, F-45071 Orléans, France.
| | - Nathalie Mignet
- CNRS, UTCBS UMR 8258, F-75006 Paris, France; Université Paris Descartes, Sorbonne-Paris-Cité, UTCBS, F-75006 Paris, France; Chimie ParisTech, PSL Research University, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), F-75005 Paris, France; INSERM, UTCBS U 1022, F-75006 Paris, France
| |
Collapse
|
31
|
Murata Y, Jo JI, Tabata Y. Preparation of gelatin nanospheres incorporating quantum dots and iron oxide nanoparticles for multimodal cell imaging. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2017; 28:555-568. [PMID: 28142329 DOI: 10.1080/09205063.2017.1286185] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The objective of this study is to prepare a multimodal probe which can simultaneously visualize cells by optical and magnetic resonance (MR) imaging modalities. Gelatin nanospheres incorporating quantum dots (QD) and iron oxide nanoparticles (IONP) were prepared by the conventional emulsion method. The percentage of QD and IONP incorporated in gelatin nanospheres was changed by the concentrations of gelatin and glutaraldehyde used. However, the apparent size and surface zeta potential were hardly changed. Gelatin nanospheres incorporating QD and IONP were treated with octa-arginine (R8) of a cell-penetrating peptide. When incubated with normal human articular chondrocytes, gelatin nanospheres incorporating QD and IONP were efficiently internalized into the cells although their cytotoxicity was observed at the R8 concentration of 320 μM. The cells internalizing gelatin nanospheres incorporating QD and IONP could be visualized by both the optical and MR imaging modalities. It is concluded that gelatin nanospheres incorporating QD and IONP are promising for the probe of multimodal cell imaging.
Collapse
Affiliation(s)
- Yuki Murata
- a Laboratory of Biomaterials, Department of Regeneration Science and Engineering , Institute for Frontier Life and Medical Sciences, Kyoto University , Kyoto , Japan
| | - Jun-Ichiro Jo
- a Laboratory of Biomaterials, Department of Regeneration Science and Engineering , Institute for Frontier Life and Medical Sciences, Kyoto University , Kyoto , Japan
| | - Yasuhiko Tabata
- a Laboratory of Biomaterials, Department of Regeneration Science and Engineering , Institute for Frontier Life and Medical Sciences, Kyoto University , Kyoto , Japan
| |
Collapse
|
32
|
Carugo D, Aron M, Sezgin E, Bernardino de la Serna J, Kuimova MK, Eggeling C, Stride E. Modulation of the molecular arrangement in artificial and biological membranes by phospholipid-shelled microbubbles. Biomaterials 2017; 113:105-117. [DOI: 10.1016/j.biomaterials.2016.10.034] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 10/22/2016] [Accepted: 10/23/2016] [Indexed: 12/17/2022]
|
33
|
Wei S, Xu C, Rychak JJ, Luong A, Sun Y, Yang Z, Li M, Liu C, Fu N, Yang B. Short Hairpin RNA Knockdown of Connective Tissue Growth Factor by Ultrasound-Targeted Microbubble Destruction Improves Renal Fibrosis. ULTRASOUND IN MEDICINE & BIOLOGY 2016; 42:2926-2937. [PMID: 27597128 DOI: 10.1016/j.ultrasmedbio.2016.07.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Revised: 07/14/2016] [Accepted: 07/25/2016] [Indexed: 06/06/2023]
Abstract
The purpose of this study was to evaluate whether ultrasound-targeted microbubble destruction transfer of interfering RNA against connective tissue growth factor (CTGF) in the kidney would ameliorate renal fibrosis in vivo. A short hairpin RNA (shRNA) targeting CTGF was cloned into a tool plasmid and loaded onto the surface of a cationic microbubble product. A unilateral ureteral obstruction (UUO) model in mice was used to evaluate the effect of CTGF knockdown. Mice were administered the plasmid-carrying microbubble intravenously, and ultrasound was applied locally to the obstructed kidney. Mice undergoing a sham UUO surgery and untreated UUO mice were used as disease controls, and mice administered plasmid alone, plasmid with ultrasound treatment and microbubbles and plasmid without ultrasound were used as treatment controls. Mice were treated once and then evaluated at day 14. CTGF in the kidney was measured by quantitative reverse transcription polymerase chain reaction and Western blot. Expression of CTGF, transforming growth factor β1, α smooth muscle actin and type I collagen in the obstructed kidney was evaluated by immunohistochemistry. The cohort treated with plasmid-carrying microbubbles and ultrasound exhibited reduced mRNA and protein expression of CTGF (p < 0.01). Furthermore, CTGF gene silencing decreased the interstitial deposition of transforming growth factor β1, α smooth muscle actin and type I collagen as assessed in immunohistochemistry, as well as reduced renal fibrosis in pathologic alterations (p < 0.01). No significant changes in target mRNA, protein expression or disease pathology were observed in the control cohorts. A single treatment of ultrasound-targeted microbubble destruction is able to deliver sufficient shRNA to inhibit the expression of CTGF and provide a meaningful reduction in disease severity. This technique may be a potential therapy for treatment of renal fibrosis.
Collapse
Affiliation(s)
- Shuping Wei
- Department of Ultrasound, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu Province, China
| | - Chaoli Xu
- Department of Ultrasound, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu Province, China
| | | | | | - Yu Sun
- Department of Pharmacological Study, Origin Biosciences, Inc., Nanjing, Jiangsu Province, China
| | - Zhijian Yang
- Department of Pharmacological Study, Origin Biosciences, Inc., Nanjing, Jiangsu Province, China
| | - Mingxia Li
- Department of Ultrasound, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu Province, China
| | - Chunrui Liu
- Department of Ultrasound, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu Province, China
| | - Ninghua Fu
- Department of Ultrasound, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu Province, China
| | - Bin Yang
- Department of Ultrasound, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
34
|
Abstract
More than 0.5 million new cases of head and neck cancer are diagnosed worldwide each year, and approximately 75% of them are treated with radiation alone or in combination with other cancer treatments. A majority of patients treated with radiotherapy develop significant oral off-target effects because of the unavoidable irradiation of normal tissues. Salivary glands that lie within treatment fields are often irreparably damaged and a decline in function manifests as dry mouth or xerostomia. Limited ability of the salivary glands to regenerate lost acinar cells makes radiation-induced loss of function a chronic problem that affects the quality of life of the patients well beyond the completion of radiotherapy. The restoration of saliva production after irradiation has been a daunting challenge, and this review provides an overview of promising gene therapeutics that either improve the gland’s ability to survive radiation insult, or alternately, restore fluid flow after radiation. The salient features and shortcomings of each approach are discussed.
Collapse
Affiliation(s)
- Renjith Parameswaran Nair
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, Louisiana 71130, United States of America
| | - Gulshan Sunavala-Dossabhoy
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, Louisiana 71130, United States of America
| |
Collapse
|
35
|
Dimcevski G, Kotopoulis S, Bjånes T, Hoem D, Schjøtt J, Gjertsen BT, Biermann M, Molven A, Sorbye H, McCormack E, Postema M, Gilja OH. A human clinical trial using ultrasound and microbubbles to enhance gemcitabine treatment of inoperable pancreatic cancer. J Control Release 2016; 243:172-181. [PMID: 27744037 DOI: 10.1016/j.jconrel.2016.10.007] [Citation(s) in RCA: 317] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 10/07/2016] [Accepted: 10/10/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND The primary aim of our study was to evaluate the safety and potential toxicity of gemcitabine combined with microbubbles under sonication in inoperable pancreatic cancer patients. The secondary aim was to evaluate a novel image-guided microbubble-based therapy, based on commercially available technology, towards improving chemotherapeutic efficacy, preserving patient performance status, and prolonging survival. METHODS Ten patients were enrolled and treated in this Phase I clinical trial. Gemcitabine was infused intravenously over 30min. Subsequently, patients were treated using a commercial clinical ultrasound scanner for 31.5min. SonoVue® was injected intravenously (0.5ml followed by 5ml saline every 3.5min) during the ultrasound treatment with the aim of inducing sonoporation, thus enhancing therapeutic efficacy. RESULTS The combined therapeutic regimen did not induce any additional toxicity or increased frequency of side effects when compared to gemcitabine chemotherapy alone (historical controls). Combination treated patients (n=10) tolerated an increased number of gemcitabine cycles compared with historical controls (n=63 patients; average of 8.3±6.0cycles, versus 13.8±5.6cycles, p=0.008, unpaired t-test). In five patients, the maximum tumour diameter was decreased from the first to last treatment. The median survival in our patients (n=10) was also increased from 8.9months to 17.6months (p=0.011). CONCLUSIONS It is possible to combine ultrasound, microbubbles, and chemotherapy in a clinical setting using commercially available equipment with no additional toxicities. This combined treatment may improve the clinical efficacy of gemcitabine, prolong the quality of life, and extend survival in patients with pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Georg Dimcevski
- National Centre for Ultrasound in Gastroenterology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway.
| | - Spiros Kotopoulis
- National Centre for Ultrasound in Gastroenterology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Tormod Bjånes
- Laboratory of Clinical Biochemistry, Section of Clinical Pharmacology, Haukeland University Hospital, Bergen, Norway
| | - Dag Hoem
- Department of Surgical Sciences, Haukeland University Hospital, Norway
| | - Jan Schjøtt
- Laboratory of Clinical Biochemistry, Section of Clinical Pharmacology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Bjørn Tore Gjertsen
- Centre for Cancer Biomarkers, CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Bergen, Norway
| | - Martin Biermann
- Department of Radiology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Anders Molven
- Department of Pathology, Haukeland University Hospital, Bergen, Norway; Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Halfdan Sorbye
- Department of Oncology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Emmet McCormack
- Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Bergen, Norway
| | - Michiel Postema
- Institute of Fundamental Technological Research, Polish Academy of Sciences, Warszawa, Poland; School of Electrical and Information Engineering, Chamber of Mines Building, University of the Witwatersrand, Johannesburg, South Africa
| | - Odd Helge Gilja
- National Centre for Ultrasound in Gastroenterology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
36
|
Xing L, Shi Q, Zheng K, Shen M, Ma J, Li F, Liu Y, Lin L, Tu W, Duan Y, Du L. Ultrasound-Mediated Microbubble Destruction (UMMD) Facilitates the Delivery of CA19-9 Targeted and Paclitaxel Loaded mPEG-PLGA-PLL Nanoparticles in Pancreatic Cancer. Am J Cancer Res 2016; 6:1573-87. [PMID: 27446491 PMCID: PMC4955056 DOI: 10.7150/thno.15164] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 05/01/2016] [Indexed: 02/07/2023] Open
Abstract
Pancreatic cancer, one of the most lethal human malignancies with dismal prognosis, is refractory to existing radio-chemotherapeutic treatment modalities. There is a critical unmet need to develop effective approaches, especially for targeted pancreatic cancer drug delivery. Targeted and drug-loaded nanoparticles (NPs) combined with ultrasound-mediated microbubble destruction (UMMD) have been shown to significantly increase the cellular uptake in vitro and drug retention in vivo, suggesting a promising strategy for cancer therapy. In this study, we synthesized pancreatic cancer-targeting organic NPs that were modified with anti CA19-9 antibody and encapsulated paclitaxol (PTX). The three-block copolymer methoxy polyethylene glycol-polylacticco-glycolic acid-polylysine (mPEG-PLGA-PLL) constituted the skeleton of the NPs. We speculated that the PTX-NPs-anti CA19-9 would circulate long-term in vivo, "actively target" pancreatic cancer cells, and sustainably release the loaded PTX while UMMD would "passively target" the irradiated tumor and effectively increase the permeability of cell membrane and capillary gaps. Our results demonstrated that the combination of PTX-NPs-anti CA19-9 with UMMD achieved a low IC50, significant cell cycle arrest, and cell apoptosis in vitro. In mouse pancreatic tumor xenografts, the combined application of PTX-NP-anti CA19-9 NPs with UMMD attained the highest tumor inhibition rate, promoted the pharmacokinetic profile by increasing AUC, t1/2, and mean residence time (MRT), and decreased clearance. Consequently, the survival of the tumor-bearing nude mice was prolonged without obvious toxicity. The dynamic change in cellular uptake, targeted real-time imaging, and the concentration of PTX in the plasma and tumor were all closely associated with the treatment efficacy both in vitro and in vivo. Our study suggests that PTX-NP-anti CA19-9 NPs combined with UMMD is a promising strategy for the treatment of pancreatic cancer.
Collapse
|
37
|
Chow YT, Chen S, Wang R, Liu C, Kong CW, Li RA, Cheng SH, Sun D. Single Cell Transfection through Precise Microinjection with Quantitatively Controlled Injection Volumes. Sci Rep 2016; 6:24127. [PMID: 27067121 PMCID: PMC4828701 DOI: 10.1038/srep24127] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 03/21/2016] [Indexed: 11/10/2022] Open
Abstract
Cell transfection is a technique wherein foreign genetic molecules are delivered into cells. To elucidate distinct responses during cell genetic modification, methods to achieve transfection at the single-cell level are of great value. Herein, we developed an automated micropipette-based quantitative microinjection technology that can deliver precise amounts of materials into cells. The developed microinjection system achieved precise single-cell microinjection by pre-patterning cells in an array and controlling the amount of substance delivered based on injection pressure and time. The precision of the proposed injection technique was examined by comparing the fluorescence intensities of fluorescent dye droplets with a standard concentration and water droplets with a known injection amount of the dye in oil. Injection of synthetic modified mRNA (modRNA) encoding green fluorescence proteins or a cocktail of plasmids encoding green and red fluorescence proteins into human foreskin fibroblast cells demonstrated that the resulting green fluorescence intensity or green/red fluorescence intensity ratio were well correlated with the amount of genetic material injected into the cells. Single-cell transfection via the developed microinjection technique will be of particular use in cases where cell transfection is challenging and genetically modified of selected cells are desired.
Collapse
Affiliation(s)
- Yu Ting Chow
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Shuxun Chen
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Ran Wang
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Chichi Liu
- Department of Biomedical Science, City University of Hong Kong, Hong Kong, China
| | - Chi-wing Kong
- Stem Cell and Regenerative Medicine Consortium, Department of Physiology, LKS Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Ronald A. Li
- Stem Cell and Regenerative Medicine Consortium, Department of Physiology, LKS Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Shuk Han Cheng
- Department of Biomedical Science, City University of Hong Kong, Hong Kong, China
| | - Dong Sun
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| |
Collapse
|
38
|
Mozafari M, Shimoda M, Urbanska AM, Laurent S. Ultrasound-targeted microbubble destruction: toward a new strategy for diabetes treatment. Drug Discov Today 2016; 21:540-3. [PMID: 26646254 DOI: 10.1016/j.drudis.2015.11.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 11/02/2015] [Accepted: 11/23/2015] [Indexed: 02/06/2023]
|
39
|
Chang L, Hu J, Chen F, Chen Z, Shi J, Yang Z, Li Y, Lee LJ. Nanoscale bio-platforms for living cell interrogation: current status and future perspectives. NANOSCALE 2016; 8:3181-3206. [PMID: 26745513 DOI: 10.1039/c5nr06694h] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The living cell is a complex entity that dynamically responds to both intracellular and extracellular environments. Extensive efforts have been devoted to the understanding intracellular functions orchestrated with mRNAs and proteins in investigation of the fate of a single-cell, including proliferation, apoptosis, motility, differentiation and mutations. The rapid development of modern cellular analysis techniques (e.g. PCR, western blotting, immunochemistry, etc.) offers new opportunities in quantitative analysis of RNA/protein expression up to a single cell level. The recent entries of nanoscale platforms that include kinds of methodologies with high spatial and temporal resolution have been widely employed to probe the living cells. In this tutorial review paper, we give insight into background introduction and technical innovation of currently reported nanoscale platforms for living cell interrogation. These highlighted technologies are documented in details within four categories, including nano-biosensors for label-free detection of living cells, nanodevices for living cell probing by intracellular marker delivery, high-throughput platforms towards clinical current, and the progress of microscopic imaging platforms for cell/tissue tracking in vitro and in vivo. Perspectives for system improvement were also discussed to solve the limitations remains in current techniques, for the purpose of clinical use in future.
Collapse
Affiliation(s)
- Lingqian Chang
- NSF Nanoscale Science and Engineering Center (NSEC), The Ohio State University, Columbus, OH 43212, USA.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Tan JKY, Pham B, Zong Y, Perez C, Maris DO, Hemphill A, Miao CH, Matula TJ, Mourad PD, Wei H, Sellers DL, Horner PJ, Pun SH. Microbubbles and ultrasound increase intraventricular polyplex gene transfer to the brain. J Control Release 2016; 231:86-93. [PMID: 26860281 DOI: 10.1016/j.jconrel.2016.02.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 01/22/2016] [Accepted: 02/02/2016] [Indexed: 12/16/2022]
Abstract
Neurons in the brain can be damaged or lost from neurodegenerative disease, stroke, or traumatic injury. Although neurogenesis occurs in mammalian adult brains, the levels of natural neurogenesis are insufficient to restore function in these cases. Gene therapy has been pursued as a promising strategy to induce differentiation of neural progenitor cells into functional neurons. Non-viral vectors are a preferred method of gene transfer due to potential safety and manufacturing benefits but suffer from lower delivery efficiencies compared to viral vectors. Since the neural stem and progenitor cells reside in the subventricular zone of the brain, intraventricular injection has been used as an administration route for gene transfer to these cells. However, the choroid plexus epithelium remains an obstacle to delivery. Recently, transient disruption of the blood-brain barrier by microbubble-enhanced ultrasound has been used to successfully improve drug delivery to the brain after intravenous injection. In this work, we demonstrate that microbubble-enhanced ultrasound can similarly improve gene transfer to the subventricular zone after intraventricular injection. Microbubbles of different surface charges (neutral, slightly cationic, and cationic) were prepared, characterized by acoustic flow cytometry, and evaluated for their ability to increase the permeability of immortalized choroid plexus epithelium monolayers in vitro. Based on these results, slightly cationic microbubbles were evaluated for microbubble and ultrasound-mediated enhancement of non-viral gene transfer in vivo. When coupled with our previously reported gene delivery vehicles, the slightly cationic microbubbles significantly increased ultrasound-mediated transfection of the murine brain when compared to commercially available Definity® microbubbles. Temporary disruption of the choroid plexus by microbubble-enhanced ultrasound is therefore a viable way of enhancing gene delivery to the brain and merits further research.
Collapse
Affiliation(s)
- James-Kevin Y Tan
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA; Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA 98195, USA
| | - Binhan Pham
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA; Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA 98195, USA
| | - Yujin Zong
- Center for Industrial and Medical Ultrasound, University of Washington, Seattle, WA 98195, USA; Department of Biomedical Engineering, Xian Jiaotong University, Xi'an, 710049, China
| | - Camilo Perez
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA; Center for Industrial and Medical Ultrasound, University of Washington, Seattle, WA 98195, USA
| | - Don O Maris
- Department of Neurological Surgery, University of Washington, Seattle, WA 98109, USA
| | - Ashton Hemphill
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA; Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA 98195, USA
| | - Carol H Miao
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | - Thomas J Matula
- Center for Industrial and Medical Ultrasound, University of Washington, Seattle, WA 98195, USA
| | - Pierre D Mourad
- Department of Neurological Surgery, University of Washington, Seattle, WA 98109, USA
| | - Hua Wei
- Department of Chemistry, Lanzhou University, Lanzhou 730000, China
| | - Drew L Sellers
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA; Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA 98195, USA
| | - Philip J Horner
- Department of Neurological Surgery, University of Washington, Seattle, WA 98109, USA.
| | - Suzie H Pun
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA; Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
41
|
Xiang X, Tang Y, Leng Q, Zhang L, Qiu L. Targeted gene delivery to the synovial pannus in antigen-induced arthritis by ultrasound-targeted microbubble destruction in vivo. ULTRASONICS 2016; 65:304-314. [PMID: 26433434 DOI: 10.1016/j.ultras.2015.09.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 09/09/2015] [Accepted: 09/16/2015] [Indexed: 06/05/2023]
Abstract
The purpose of this study was to optimize an ultrasound-targeted microbubble destruction (UTMD) technique to improve the in vivo transfection efficiency of the gene encoding enhanced green fluorescent protein (EGFP) in the synovial pannus in an antigen-induced arthritis rabbit model. A mixture of microbubbles and plasmids was locally injected into the knee joints of an antigen-induced arthritis (AIA) rabbits. The plasmid concentrations and ultrasound conditions were varied in the experiments. We also tested local articular and intravenous injections. The rabbits were divided into five groups: (1) ultrasound+microbubbles+plasmid; (2) ultrasound+plasmid; (3) microbubble+plasmid; (4) plasmid only; (5) untreated controls. EGFP expression was observed by fluorescent microscope and immunohistochemical staining in the synovial pannus of each group. The optimal plasmid dosage and ultrasound parameter were determined based on the results of EGFP expression and the present and absent of tissue damage under light microscopy. The irradiation procedure was performed to observe the duration of the EGFP expression in the synovial pannus and other tissues and organs, as well as the damage to the normal cells. The optimal condition was determined to be a 1-MHz ultrasound pulse applied for 5 min with a power output of 2 W/cm(2) and a 20% duty cycle along with 300 μg of plasmid. Under these conditions, the synovial pannus showed significant EGFP expression without significant damage to the surrounding normal tissue. The EGFP expression induced by the local intra-articular injection was significantly more increased than that induced by the intravenous injection. The EGFP expression in the synovial pannus of the ultrasound+microbubbles+plasmid group was significantly higher than that of the other four groups (P<0.05). The expression peaked on day 5, remained detectable on day 40 and disappeared on day 60. No EGFP expression was detected in the other tissues and organs. The UTMD technique can significantly enhance the in vivo gene transfection efficiency without significant tissue damage in the synovial pannus of an AIA model. Thus, this could become a safe and effective non-viral gene transfection procedure for arthritis therapy.
Collapse
Affiliation(s)
- Xi Xiang
- Department of Ultrasound, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yuanjiao Tang
- Department of Ultrasound, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Qianying Leng
- Department of Ultrasound, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Lingyan Zhang
- Department of Ultrasound, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Li Qiu
- Department of Ultrasound, West China Hospital of Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
42
|
|