1
|
Chen L, Zhang S, Duan Y, Song X, Chang M, Feng W, Chen Y. Silicon-containing nanomedicine and biomaterials: materials chemistry, multi-dimensional design, and biomedical application. Chem Soc Rev 2024; 53:1167-1315. [PMID: 38168612 DOI: 10.1039/d1cs01022k] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
The invention of silica-based bioactive glass in the late 1960s has sparked significant interest in exploring a wide range of silicon-containing biomaterials from the macroscale to the nanoscale. Over the past few decades, these biomaterials have been extensively explored for their potential in diverse biomedical applications, considering their remarkable bioactivity, excellent biocompatibility, facile surface functionalization, controllable synthesis, etc. However, to expedite the clinical translation and the unexpected utilization of silicon-composed nanomedicine and biomaterials, it is highly desirable to achieve a thorough comprehension of their characteristics and biological effects from an overall perspective. In this review, we provide a comprehensive discussion on the state-of-the-art progress of silicon-composed biomaterials, including their classification, characteristics, fabrication methods, and versatile biomedical applications. Additionally, we highlight the multi-dimensional design of both pure and hybrid silicon-composed nanomedicine and biomaterials and their intrinsic biological effects and interactions with biological systems. Their extensive biomedical applications span from drug delivery and bioimaging to therapeutic interventions and regenerative medicine, showcasing the significance of their rational design and fabrication to meet specific requirements and optimize their theranostic performance. Additionally, we offer insights into the future prospects and potential challenges regarding silicon-composed nanomedicine and biomaterials. By shedding light on these exciting research advances, we aspire to foster further progress in the biomedical field and drive the development of innovative silicon-composed nanomedicine and biomaterials with transformative applications in biomedicine.
Collapse
Affiliation(s)
- Liang Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Shanshan Zhang
- Department of Ultrasound Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P. R. China
| | - Yanqiu Duan
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, P. R. China.
| | - Xinran Song
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Meiqi Chang
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, P. R. China.
| | - Wei Feng
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| |
Collapse
|
2
|
Cui Z, Jiao Y, Pu L, Tang JZ, Wang G. The Progress of Non-Viral Materials and Methods for Gene Delivery to Skeletal Muscle. Pharmaceutics 2022; 14:2428. [PMID: 36365246 PMCID: PMC9695315 DOI: 10.3390/pharmaceutics14112428] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/27/2022] [Accepted: 11/02/2022] [Indexed: 09/10/2024] Open
Abstract
Since Jon A. Wolff found skeletal muscle cells being able to express foreign genes and Russell J. Mumper increased the gene transfection efficiency into the myocytes by adding polymers, skeletal muscles have become a potential gene delivery and expression target. Different methods have been developing to deliver transgene into skeletal muscles. Among them, viral vectors may achieve potent gene delivery efficiency. However, the potential for triggering biosafety risks limited their clinical applications. Therefore, non-viral biomaterial-mediated methods with reliable biocompatibility are promising tools for intramuscular gene delivery in situ. In recent years, a series of advanced non-viral gene delivery materials and related methods have been reported, such as polymers, liposomes, cell penetrating peptides, as well as physical delivery methods. In this review, we summarized the research progresses and challenges in non-viral intramuscular gene delivery materials and related methods, focusing on the achievements and future directions of polymers.
Collapse
Affiliation(s)
- Zhanpeng Cui
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Yang Jiao
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Linyu Pu
- School of Materials and Chemistry, Southwest University of Science and Technology, Mianyang 621010, China
| | - James Zhenggui Tang
- Research Institute in Healthcare Science, Faculty of Science & Engineering, University of Wolverhampton, Wolverhampton WV1 1SB, UK
| | - Gang Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| |
Collapse
|
3
|
Abou-Saleh RH, McLaughlan JR, Bushby RJ, Johnson BR, Freear S, Evans SD, Thomson NH. Molecular Effects of Glycerol on Lipid Monolayers at the Gas-Liquid Interface: Impact on Microbubble Physical and Mechanical Properties. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:10097-10105. [PMID: 30901226 DOI: 10.1021/acs.langmuir.8b04130] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
The production and stability of microbubbles (MBs) is enhanced by increasing the viscosity of both the formation and storage solution, respectively. Glycerol is a good candidate for biomedical applications of MBs, since it is biocompatible, although the exact molecular mechanisms of its action is not fully understood. Here, we investigate the influence glycerol has on lipid-shelled MB properties, using a range of techniques. Population lifetime and single bubble stability were studied using optical microscopy. Bubble stiffness measured by AFM compression is compared with lipid monolayer behavior in a Langmuir-Blodgett trough. We deduce that increasing glycerol concentrations enhances stability of MB populations through a 3-fold mechanism. First, binding of glycerol to lipid headgroups in the interfacial monolayer up to 10% glycerol increases MB stiffness but has limited impact on shell resistance to gas permeation and corresponding MB lifetime. Second, increased solution viscosity above 10% glycerol slows down the kinetics of gas transfer, markedly increasing MB stability. Third, above 10%, glycerol induces water structuring around the lipid monolayer, forming a glassy layer which also increases MB stiffness and resistance to gas loss. At 30% glycerol, the glassy layer is ablated, lowering the MB stiffness, but MB stability is further augmented. Although the molecular interactions of glycerol with the lipid monolayer modulate the MB lipid shell properties, MB lifetime continually increases from 0 to 30% glycerol, indicating that its viscosity is the dominant effect on MB solution stability. This three-fold action and biocompatibility makes glycerol ideal for therapeutic MB formation and storage and gives new insight into the action of glycerol on lipid monolayers at the gas-liquid interface.
Collapse
Affiliation(s)
- Radwa H Abou-Saleh
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy , University of Leeds , Leeds LS2 9JT , United Kingdom
- Biophysics Group, Department of Physics, Faculty of Science , Mansoura University , Mansoura , Egypt
| | - James R McLaughlan
- School of Electronic and Electrical Engineering , University of Leeds , Leeds LS2 9JT , United Kingdom
- Leeds Institute of Medical Research , University of Leeds, St. James's University Hospital , Leeds LS9 7TF , United Kingdom
| | - Richard J Bushby
- School of Chemistry , University of Leeds , Leeds LS2 9JT , United Kingdom
| | - Benjamin R Johnson
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy , University of Leeds , Leeds LS2 9JT , United Kingdom
| | - Steven Freear
- School of Electronic and Electrical Engineering , University of Leeds , Leeds LS2 9JT , United Kingdom
| | - Stephen D Evans
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy , University of Leeds , Leeds LS2 9JT , United Kingdom
| | - Neil H Thomson
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy , University of Leeds , Leeds LS2 9JT , United Kingdom
- Division of Oral Biology, School of Dentistry , University of Leeds , Leeds LS2 9LU , United Kingdom
| |
Collapse
|
4
|
Yu L, Lin H, Lu X, Chen Y. Multifunctional Mesoporous Silica Nanoprobes: Material Chemistry–Based Fabrication and Bio‐Imaging Functionality. ADVANCED THERAPEUTICS 2018; 1. [DOI: 10.1002/adtp.201800078] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Indexed: 12/25/2022]
Abstract
AbstractNanoparticles‐based bioimaging probes are attracting broad attention for various biomedical applications. As one of the mostly explored nanoplatforms, mesoporous silica nanoparticles (MSNs) show high clinical‐translation potential for diagnostic probing/imaging. Based on their tunable morphology, abundant surface chemistry, and well‐defined mesostructure, MSNs are regarded as the desirable platforms for constructing diverse nanoprobes via incorporation of a variety of functional moieties or components. In this review, the authors summarize and discuss recent progress in the rational design and fabrication of multifunctional mesoporous silica‐based composite nanoprobes for versatile bioimaging applications. Four kinds of methodologies for the fabrication of these mesoporous silica‐based nanoprobes are discussed, including encapsulating functional nanoparticles within a mesoporous silica shell, assembling functional nanoparticles on the surface of MSNs, dispersing nanoparticles into the nanometer‐scale mesopores of MSNs, and doping functional moieties into the framework of MSNs. The applications of mesoporous silica nanoprobes in magnetic resonance imaging, ultrasound imaging, computed tomography imaging, fluorescence imaging, positron emission computed tomography, photoacoustic (PA) imaging, and even multimodality imaging are discussed in detail. The biosafety of MSN‐based composite nanoplatforms as bioimaging nanoprobes is also highlighted, accompanied by a deep discussion on facing the challenges and future developments for guaranteeing their further potential clinical translation.
Collapse
Affiliation(s)
- Luodan Yu
- State Key Laboratory of High Performance Ceramic and Superfine Microstructures Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
- University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Han Lin
- State Key Laboratory of High Performance Ceramic and Superfine Microstructures Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
- University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Xiangyu Lu
- State Key Laboratory of High Performance Ceramic and Superfine Microstructures Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
- University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Yu Chen
- State Key Laboratory of High Performance Ceramic and Superfine Microstructures Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
| |
Collapse
|
5
|
Tang H, Zheng Y, Chen Y. Materials Chemistry of Nanoultrasonic Biomedicine. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29:1604105. [PMID: 27991697 DOI: 10.1002/adma.201604105] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 10/12/2016] [Indexed: 06/06/2023]
Abstract
As a special cross-disciplinary research frontier, nanoultrasonic biomedicine refers to the design and synthesis of nanomaterials to solve some critical issues of ultrasound (US)-based biomedicine. The concept of nanoultrasonic biomedicine can also overcome the drawbacks of traditional microbubbles and promote the generation of novel US-based contrast agents or synergistic agents for US theranostics. Here, we discuss the recent developments of material chemistry in advancing the nanoultrasonic biomedicine for diverse US-based bio-applications. We initially introduce the design principles of novel nanoplatforms for serving the nanoultrasonic biomedicine, from the viewpoint of synthetic material chemistry. Based on these principles and diverse US-based bio-application backgrounds, the representative proof-of-concept paradigms on this topic are clarified in detail, including nanodroplet vaporization for intelligent/responsive US imaging, multifunctional nano-contrast agents for US-based multi-modality imaging, activatable synergistic agents for US-based therapy, US-triggered on-demand drug releasing, US-enhanced gene transfection, US-based synergistic therapy on combating the cancer and potential toxicity issue of screening various nanosystems suitable for nanoultrasonic biomedicine. It is highly expected that this novel nanoultrasonic biomedicine and corresponding high performance in US imaging and therapy can significantly promote the generation of new sub-discipline of US-based biomedicine by rationally integrating material chemistry and theranostic nanomedicine with clinical US-based biomedicine.
Collapse
Affiliation(s)
- Hailin Tang
- Department of Diagnostic Ultrasound, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, P. R. China
| | - Yuanyi Zheng
- Shanghai Institute of Ultrasound in Medicine, Shanghai Jiaotong University Affiliated, Shanghai Sixth People's Hospital, Shanghai, 200233, P. R. China
| | - Yu Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| |
Collapse
|
6
|
Chen HH, Matkar PN, Afrasiabi K, Kuliszewski MA, Leong-Poi H. Prospect of ultrasound-mediated gene delivery in cardiovascular applications. Expert Opin Biol Ther 2016; 16:815-26. [DOI: 10.1517/14712598.2016.1169268] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
7
|
Hsiang YH, Song J, Price RJ. The partitioning of nanoparticles to endothelium or interstitium during ultrasound-microbubble-targeted delivery depends on peak-negative pressure. JOURNAL OF NANOPARTICLE RESEARCH : AN INTERDISCIPLINARY FORUM FOR NANOSCALE SCIENCE AND TECHNOLOGY 2015; 17:345. [PMID: 26594129 PMCID: PMC4651175 DOI: 10.1007/s11051-015-3153-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 08/13/2015] [Indexed: 06/05/2023]
Abstract
Patients diagnosed with advanced peripheral arterial disease often face poor prognoses and have limited treatment options. For some patient populations, the therapeutic growth of collateral arteries (i.e. arteriogenesis) that bypass regions affected by vascular disease may become a viable treatment option. Our group and others are developing therapeutic approaches centered on the ability of ultrasound-activated microbubbles to permeabilize skeletal muscle capillaries and facilitate the targeted delivery of pro-arteriogenic growth factor-bearing nanoparticles. The development of such approaches would benefit significantly from a better understanding of how nanoparticle diameter and ultrasound peak-negative pressure affect both total nanoparticle delivery and the partitioning of nanoparticles to endothelial or interstitial compartments. Toward this goal, using Balb/C mice that had undergone unilateral femoral artery ligation, we intra-arterially co-injected nanoparticles (50 and 100 nm) with microbubbles, applied 1 MHz ultrasound to the gracilis adductor muscle at peak-negative pressures of 0.7, 0.55, 0.4, and 0.2 MPa, and analyzed nanoparticle delivery and distribution. As expected, total nanoparticle (50 and 100 nm) delivery increased with increasing peak-negative pressure, with 50 nm nanoparticles exhibiting greater tissue coverage than 100 nm nanoparticles. Of particular interest, increasing peak-negative pressure resulted in increased delivery to the interstitium for both nanoparticle sizes, but had little influence on nanoparticle delivery to the endothelium. Thus, we conclude that alterations to peak-negative pressure may be used to adjust the fraction of nanoparticles delivered to the interstitial compartment. This information will be useful when designing ultrasound protocols for delivering pro-arteriogenic nanoparticles to skeletal muscle.
Collapse
Affiliation(s)
- Y.-H. Hsiang
- Department of Biomedical Engineering, University of Virginia, Box 800759, Health System, Charlottesville, VA 22908, USA
| | - J. Song
- Department of Biomedical Engineering, University of Virginia, Box 800759, Health System, Charlottesville, VA 22908, USA
| | - R. J. Price
- Department of Biomedical Engineering, University of Virginia, Box 800759, Health System, Charlottesville, VA 22908, USA
| |
Collapse
|
8
|
Wan C, Li F, Li H. Gene therapy for ocular diseases meditated by ultrasound and microbubbles (Review). Mol Med Rep 2015; 12:4803-14. [PMID: 26151686 PMCID: PMC4581786 DOI: 10.3892/mmr.2015.4054] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 06/03/2015] [Indexed: 02/06/2023] Open
Abstract
The eye is an ideal target organ for gene therapy as it is easily accessible and immune‑privileged. With the increasing insight into the underlying molecular mechanisms of ocular diseases, gene therapy has been proposed as an effective approach. Successful gene therapy depends on efficient gene transfer to targeted cells to prove stable and prolonged gene expression with minimal toxicity. At present, the main hindrance regarding the clinical application of gene therapy is not the lack of an ideal gene, but rather the lack of a safe and efficient method to selectively deliver genes to target cells and tissues. Ultrasound‑targeted microbubble destruction (UTMD), with the advantages of high safety, repetitive applicability and tissue targeting, has become a potential strategy for gene‑ and drug delivery. When gene‑loaded microbubbles are injected, UTMD is able to enhance the transport of the gene to the targeted cells. High‑amplitude oscillations of microbubbles act as cavitation nuclei which can effectively focus ultrasound energy, produce oscillations and disruptions that increase the permeability of the cell membrane and create transient pores in the cell membrane. Thereby, the efficiency of gene therapy can be significantly improved. The UTMD‑mediated gene delivery system has been widely used in pre‑clinical studies to enhance gene expression in a site‑specific manner in a variety of organs. With reasonable application, the effects of sonoporation can be spatially and temporally controlled to improve localized tissue deposition of gene complexes for ocular gene therapy applications. In addition, appropriately powered, focused ultrasound combined with microbubbles can induce a reversible disruption of the blood‑retinal barrier with no significant side effects. The present review discusses the current status of gene therapy of ocular diseases as well as studies on gene therapy of ocular diseases meditated by UTMD.
Collapse
Affiliation(s)
- Caifeng Wan
- Department of Ultrasound, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Fenghua Li
- Department of Ultrasound, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Hongli Li
- Department of Ultrasound, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| |
Collapse
|
9
|
Wan C, Qian J, Li F, Li H. Ultrasound-targeted microbubble destruction enhances polyethylenimine-mediated gene transfection in vitro in human retinal pigment epithelial cells and in vivo in rat retina. Mol Med Rep 2015; 12:2835-41. [PMID: 25936880 DOI: 10.3892/mmr.2015.3703] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 03/16/2015] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate the efficacy and safety of ultrasound-targeted microbubble destruction (UTMD)-mediated polyethylenimine (PEI) transfection in cultured human retinal pigment epithelial (RPE) cells in vitro and rat retinas in vivo. An enhanced green fluorescent protein plasmid (pEGFP) was incubated with PEI to prepare a cationic complex (PEI/pEGFP), which was confirmed using a gel retardation assay. In the in vitro study, cultured human RPE cells were subjected to US waves under different conditions with or without microbubbles. The effect of UTMD on the viability of the cells was evaluated. In the in vivo study, gene transfer was examined by injecting PEI/pEGFP into the subretinal space of the rats. The rats treated with PEI/pEGFP and UTMD served as the experimental group, while rats treated with PEI/pEGFP alone served as the control group. The transfected tissue was visualized using an inverted fluorescence microscope. The expression of EGFP was classified into three groups, negative, weak positive and strong positive. Hematoxylin and eosin staining of frozen sections was used to observe tissue damage and the location of the EGFP gene expression. The electrophoresis experiment revealed that PEI treatment was able to condense DNA efficiently. In the in vitro study, the gene transfer efficiency under the optimal UTMD condition was enhanced and significantly higher than control groups. In the in vivo study, UTMD was able to enhance transgene expression in the retina without marked tissue damage. Frozen sections of the optic cups exhibited pEGFP‑positive cells, predominantly distributed in the retina. This noninvasive novel combination of UTMD with PEI was able to enhance targeted gene delivery and gene expression in the rat retina without causing any apparent tissue damage, and may be a safe method to transfer genes and drug treatments directly to the retina, therefore being of potential therapeutic value.
Collapse
Affiliation(s)
- Caifeng Wan
- Department of Ultrasound, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Jin Qian
- Department of Ophthalmology, The Baoshan Branch Institute of Shanghai Shuguang Hospital, Shanghai 201900, P.R. China
| | - Fenghua Li
- Department of Ultrasound, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Hongli Li
- Department of Ultrasound, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| |
Collapse
|
10
|
Gao R, Zhou X, Yang Y, Wang Z. Transfection of wtp53 and Rb94 genes into retinoblastomas of nude mice by ultrasound-targeted microbubble destruction. ULTRASOUND IN MEDICINE & BIOLOGY 2014; 40:2662-2670. [PMID: 25218456 DOI: 10.1016/j.ultrasmedbio.2014.05.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 05/09/2014] [Accepted: 05/19/2014] [Indexed: 06/03/2023]
Abstract
Using ultrasound-targeted microbubble destruction (UTMD), we transfected both wild-type p53 (wtp53) and Rb94 genes into retinoblastomas (RBs) of nude mice to investigate the inhibitory role of these two genes in RB development. The 40 tumor-bearing mice, which had been established by sub-retinal injection of an HXO-Rb44 cell suspension, were randomly divided into five groups: blank control group (C); blank plasmid group (M); wtp53 plasmid group (p53); Rb94 plasmid group (Rb94); wtp53 + Rb94 plasmid group (p53 + Rb94). For preparation of the DNA-loaded microbubbles, a pre-determined amount of blank plasmid, pVIVO1-p53, pVIVO1-Rb94 or pVIVO1-p53-Rb94 was added and mixed with the microbubbles. Then, these DNA-loaded microbubbles were respectively transfected into the animal model by UTMD. Vascular endothelial growth factor level and microvessel density of the tumor were determined by immunohistochemical staining. Apoptosis of tissues was detected by terminal deoxynucleotidyl transferase dUTP nick end labeling staining. Expression of wtp53 and Rb94 at both the gene and protein levels was detected by RT-PCR (reverse transcription polymerase chain reaction) and Western blot, respectively. Transfection of both genes had greater inhibitory effects on RB development and resulted in lower levels of vascular endothelial growth factor, lower microvessel density and more obvious apoptosis than single-gene transfection (p < 0.05). The results indicate that the transfection of both genes into the RB by UTMD more strongly inhibited RB growth than transfection of a single gene.
Collapse
Affiliation(s)
- Ruiqi Gao
- Department of Ophthalmology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China; Department of Ophthalmology, Dujiangyan Medical Center, Sichuan, China
| | - Xiyuan Zhou
- Department of Ophthalmology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Yingxue Yang
- Department of Ophthalmology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhigang Wang
- Institute of Ultrasonic Imaging, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Ophthalmology, Chongqing, China
| |
Collapse
|
11
|
Rychak JJ, Klibanov AL. Nucleic acid delivery with microbubbles and ultrasound. Adv Drug Deliv Rev 2014; 72:82-93. [PMID: 24486388 PMCID: PMC4204336 DOI: 10.1016/j.addr.2014.01.009] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Revised: 01/20/2014] [Accepted: 01/23/2014] [Indexed: 02/02/2023]
Abstract
Nucleic acid-based therapy is a growing field of drug delivery research. Although ultrasound has been suggested to enhance transfection decades ago, it took a combination of ultrasound with nucleic acid carrier systems (microbubbles, liposomes, polyplexes, and viral carriers) to achieve reasonable nucleic acid delivery efficacy. Microbubbles serve as foci for local deposition of ultrasound energy near the target cell, and greatly enhance sonoporation. The major advantage of this approach is in the minimal transfection in the non-insonated non-target tissues. Microbubbles can be simply co-administered with the nucleic acid carrier or can be modified to carry nucleic acid themselves. Liposomes with embedded gas or gas precursor particles can also be used to carry nucleic acid, release and deliver it by the ultrasound trigger. Successful testing in a wide variety of animal models (myocardium, solid tumors, skeletal muscle, and pancreas) proves the potential usefulness of this technique for nucleic acid drug delivery.
Collapse
Affiliation(s)
| | - Alexander L Klibanov
- Cardiovascular Division, University of Virginia, Charlottesville, VA 22908-1394, USA.
| |
Collapse
|
12
|
Sorace AG, Warram JM, Mahoney M, Zinn KR, Hoyt K. Enhancement of adenovirus delivery after ultrasound-stimulated therapy in a cancer model. ULTRASOUND IN MEDICINE & BIOLOGY 2013; 39:2374-81. [PMID: 24063960 PMCID: PMC4006627 DOI: 10.1016/j.ultrasmedbio.2013.07.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 07/29/2013] [Accepted: 07/30/2013] [Indexed: 05/06/2023]
Abstract
Improving the efficiency of adenovirus (Ad) delivery to target tissues has the potential to advance the translation of cancer gene therapy. Ultrasound (US)-stimulated therapy uses microbubbles (MBs) exposed to low-intensity US energy to improve localized delivery. We hypothesize that US-stimulated gene therapy can improve Ad infection in a primary prostate tumor through enhanced tumor uptake and retention of the Ad vector. In vitro studies were performed to analyze the degree of Ad infectivity after application of US-stimulated gene therapy. A luciferase-based Ad on a ubiquitous cytomegalovirus (CMV) promoter (Ad5/3-CMV-Luc) was used in an animal model of prostate cancer (bilateral tumor growth) to evaluate Ad transduction efficiency after US-stimulated therapy. Bioluminescence imaging was employed for in vivo analysis to quantify Ad infection within the tumor. In vitro studies revealed no difference in Ad transduction between groups receiving US-stimulated therapy using high, low or sham US intensity exposures at various multiplicities of infection (MOIs) (p = 0.80). In vivo results indicated that tumors receiving US-stimulated therapy after intra-tumoral injection of Ad5/3-CMV-Luc (1 × 10(6) plaque-forming units) exhibited a 95.1% enhancement in tumor delivery compared with control tumors receiving sham US (p = 0.03). US-stimulated therapy has significant potential to immediately affect Ad-based cancer gene therapy by improving virus bioavailability in target tissues.
Collapse
Affiliation(s)
- Anna G. Sorace
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jason M. Warram
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Marshall Mahoney
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kurt R. Zinn
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Electrical & Computer Engineering, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kenneth Hoyt
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Electrical & Computer Engineering, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
13
|
The feasibility and safety of transendocardial gene injection in canine using a multifunctional intracardiac echocardiography catheter. Int J Cardiol 2013; 165:488-93. [DOI: 10.1016/j.ijcard.2011.09.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Revised: 07/08/2011] [Accepted: 09/05/2011] [Indexed: 11/22/2022]
|
14
|
Vu V, Liu Y, Sen S, Xu A, Sweeney G. Delivery of adiponectin gene to skeletal muscle using ultrasound targeted microbubbles improves insulin sensitivity and whole body glucose homeostasis. Am J Physiol Endocrinol Metab 2013; 304:E168-75. [PMID: 23132298 PMCID: PMC3543570 DOI: 10.1152/ajpendo.00493.2012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Numerous studies have shown that adiponectin confers antidiabetic effects via both insulin-like and insulin-sensitizing actions. The majority of adiponectin in circulation is derived from adipocytes; however, other tissues such as skeletal muscle can produce adiponectin. This study was designed to investigate the functional significance of adiponectin produced by skeletal muscle. We encapsulated the adiponectin gene in lipid-coated microspheres filled with octafluoropropane gas that were injected into the systemic circulation and destroyed within the microvasculature of skeletal muscle using ultrasound. We first demonstrated safe and successful targeting of luciferase and green fluorescent protein reporter genes to skeletal muscle using this approach and then confirmed efficient overexpression of adiponectin mRNA and oligomeric protein forms. Glucose tolerance test indicated that overexpression of adiponectin in skeletal muscle was able to improve glucose intolerance induced by feeding mice a high-fat diet (HFD), and this correlated with improved skeletal muscle insulin signaling. We then performed hyperinsulinemic-euglycemic clamp studies and demonstrated that adiponectin overexpression attenuated the decreases in glucose infusion rate, glucose disposal, and increase in glucose appearance induced by HFD. Ultrasound-targeted microbubble destruction (UTMD) delivery of adiponectin to skeletal muscle also enhanced serum adiponectin levels and improved hepatic insulin sensitivity. In conclusion, our data show that UTMD efficiently delivers adiponectin to skeletal muscle and that this improves insulin sensitivity and glucose homeostasis.
Collapse
Affiliation(s)
- Vivian Vu
- Department of Biology, York University, Toronto, Canada
| | | | | | | | | |
Collapse
|
15
|
Sirsi SR, Borden MA. Advances in ultrasound mediated gene therapy using microbubble contrast agents. Am J Cancer Res 2012; 2:1208-22. [PMID: 23382777 PMCID: PMC3563148 DOI: 10.7150/thno.4306] [Citation(s) in RCA: 160] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Accepted: 07/01/2012] [Indexed: 12/19/2022] Open
Abstract
Microbubble ultrasound contrast agents have the potential to dramatically improve gene therapy treatments by enhancing the delivery of therapeutic DNA to malignant tissue. The physical response of microbubbles in an ultrasound field can mechanically perturb blood vessel walls and cell membranes, enhancing drug permeability into malignant tissue. In this review, we discuss literature that provided evidence of specific mechanisms that enhance in vivo gene delivery utilizing microbubble contrast agents, namely their ability to 1) improving cell membrane permeability, 2) modulate vascular permeability, and 3) enhance endocytotic uptake in cells. Additionally, we review novel microbubble vectors that are being developed in order to exploit these mechanisms and deliver higher gene payloads with greater target specificity. Finally, we discuss some future considerations that should be addressed in the development of next-generation microbubbles in order to improve in vivo microbubble gene delivery. Overall, microbubbles are rapidly gaining popularity as efficient gene carriers, and combined with their functionality as imaging contrast agents, they represent powerful theranostic tools for image guided gene therapy applications.
Collapse
|
16
|
Zhou SJ, Li SW, Wang JJ, Liu ZJ, Yin GB, Gong JP, Liu CA. High-intensity focused ultrasound combined with herpes simplex virus thymidine kinase gene-loaded ultrasound-targeted microbubbles improved the survival of rabbits with VX2 liver tumor. J Gene Med 2012; 14:570-9. [PMID: 22941868 DOI: 10.1002/jgm.2668] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Affiliation(s)
- Shi-Ji Zhou
- Department of Hepatobiliary Surgery; Second Affiliated Hospital of Chongqing Medical University; Chongqing; China
| | - Sheng-Wei Li
- Department of Hepatobiliary Surgery; Second Affiliated Hospital of Chongqing Medical University; Chongqing; China
| | - Ji-Jian Wang
- Department of General Surgery; Second Affiliated Hospital of Chongqing Medical University; Chongqing; China
| | - Zuo-Jin Liu
- Department of Hepatobiliary Surgery; Second Affiliated Hospital of Chongqing Medical University; Chongqing; China
| | - Guo-Bing Yin
- Department of General Surgery; Second Affiliated Hospital of Chongqing Medical University; Chongqing; China
| | - Jian-Ping Gong
- Department of Hepatobiliary Surgery; Second Affiliated Hospital of Chongqing Medical University; Chongqing; China
| | - Chang-An Liu
- Department of Hepatobiliary Surgery; Second Affiliated Hospital of Chongqing Medical University; Chongqing; China
| |
Collapse
|
17
|
Direct and Doppler angle-independent measurement of blood flow velocity in small-diameter vessels using ultrasound microbubbles. Clin Imaging 2012; 36:577-83. [DOI: 10.1016/j.clinimag.2012.01.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Revised: 12/28/2011] [Accepted: 01/20/2012] [Indexed: 11/20/2022]
|
18
|
Burke CW, Suk JS, Kim AJ, Hsiang YHJ, Klibanov AL, Hanes J, Price RJ. Markedly enhanced skeletal muscle transfection achieved by the ultrasound-targeted delivery of non-viral gene nanocarriers with microbubbles. J Control Release 2012; 162:414-21. [PMID: 22800583 DOI: 10.1016/j.jconrel.2012.07.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 07/07/2012] [Indexed: 10/28/2022]
Abstract
Our goal was to enhance ultrasound (US)-targeted skeletal muscle transfection through the use of poly(ethyleneglycol) (PEG)/polyethylenimine (PEI) nanocomplex gene carriers and adjustments to US and microbubble (MB) parameters. C57BL/6 mice received an intravenous infusion of MBs and either "naked" luciferase plasmid or luciferase plasmid condensed in PEG/PEI nanocomplexes. Pulsed ultrasound (1 MHz; 0.6 MPa or 0.8 MPa) was applied to the right hindlimb for 12 min. Luciferase activity in both hindlimbs was assessed at 3, 5, 7, and 10 days post-treatment by bioluminescent imaging. When targeted to hindlimb using unsorted MBs and 0.6 MPa US, 7 days after treatment, we observed a >60-fold increase in luciferase activity in PEG/PEI nanocomplex-treated muscles over muscles treated with "naked" plasmid DNA. Luciferase activity was consistently greater after treatment with PEG/PEI nanocomplexes at 0.6 MPa as compared to 0.8 MPa. The combination of small diameter MBs and 0.6 MPa US also resulted in significantly greater gene expression when compared to concentration matched intramuscular injections, a control condition in which considerably more PEG/PEI nanocomplexes were present in tissue. This result suggests that, in addition to facilitating PEG/PEI nanocomplex delivery from the bloodstream to tissue, US enhances transfection via one or more secondary mechanisms, including increased cellular uptake and/or trafficking to the nucleus of PEG/PEI nanocomplexes. We conclude that PEG/PEI nanocomplexes may be used to markedly enhance the amplitude of US-MB-targeted skeletal muscle transfection and that activating "small" MBs with a moderate level (0.6 MPa) of acoustic pressure can further enhance these effects.
Collapse
Affiliation(s)
- Caitlin W Burke
- Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Huang Q, Deng J, Xie Z, Wang F, Chen S, Lei B, Liao P, Huang N, Wang Z, Wang Z, Cheng Y. Effective gene transfer into central nervous system following ultrasound-microbubbles-induced opening of the blood-brain barrier. ULTRASOUND IN MEDICINE & BIOLOGY 2012; 38:1234-1243. [PMID: 22677255 DOI: 10.1016/j.ultrasmedbio.2012.02.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Revised: 02/16/2012] [Accepted: 02/20/2012] [Indexed: 06/01/2023]
Abstract
To investigate whether ultrasound-targeted microbubble destruction (UTMD) could transfer gene into central nervous system (CNS) following blood-brain barrier disruption (BBBD), DNA-loaded microbubbles were infused into the mice intravenously following ultrasonic exposure. Opening of the BBB, changes of mRNA and expression of enhanced green fluorescent protein (EGFP), and safety evaluation were measured. By UTMD, EGFP were substantially expressed in the cytoplasm of the neurons at the sonicated area with minor erythrocytes extravasation and the mRNA and expression of EGFP were markedly enhanced by about 15-fold and 10-fold, respectively, than that with US alone (p < 0.01). No EGFP was detected in the mice treated with DNA-loaded microbubbles or plasmid alone. The gene expression reached a climax at 48 h, gradually reduced to a much lower level thereafter. These results demonstrated UTMD could effectively enhance exogenous gene delivery and expression in CNS following BBBD, and this technique may provide a new method for CNS gene therapy.
Collapse
Affiliation(s)
- Qin Huang
- Department of Neurosurgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Zheng MM, Zhou XY, Wang LP, Wang ZG. Experimental research of RB94 gene transfection into retinoblastoma cells using ultrasound-targeted microbubble destruction. ULTRASOUND IN MEDICINE & BIOLOGY 2012; 38:1058-1066. [PMID: 22502879 DOI: 10.1016/j.ultrasmedbio.2012.02.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Revised: 01/05/2012] [Accepted: 02/08/2012] [Indexed: 05/31/2023]
Abstract
The purpose of this study was to explore the transfection of the recombinant expression plasmid pEGFP-C1/RB94 into human retinoblastoma cells (HXO-Rb44) using ultrasound-targeted microbubble destruction (UTMD). pEGFP-C1/RB94 was transfected into HXO-Rb44 in vitro by UTMD, with liposome as the positive control. After 24 to 72 h, the expression of the reporter gene enhanced green fluorescent protein (EGFP) was observed using fluorescent microscopy and flow cytometry. The cell viability of HXO-Rb44 was measured by a MTT assay. The mRNA and proteins of RB94, caspase-3 and Bax were analyzed by reverse transcription polymerase chain reaction (RT-PCR) and Western blot. Moreover, the apoptosis rate and cell cycle progression of the cells were detected by flow cytometry. This study demonstrated that UTMD can enhance the transfection efficiency of RB94, which has an obvious impact on the inhibition of the growth process of retinoblastoma cells, suggesting that the combination of UTMD and RB94 compounds might be a useful tool for use in the gene therapy of retinoblastoma.
Collapse
Affiliation(s)
- Min-Ming Zheng
- Department of Ophthalmology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P R China
| | | | | | | |
Collapse
|
21
|
Chen WJ, Xiong ZA, Tang Y, Dong PT, Li P, Wang ZG. Feasibility and effect of ultrasound microbubble-mediated wild-type p53 gene transfection of HeLa cells. Exp Ther Med 2012; 3:999-1004. [PMID: 22970006 DOI: 10.3892/etm.2012.528] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Accepted: 03/20/2012] [Indexed: 11/06/2022] Open
Abstract
Gene therapy holds great promise for the treatment of diseases. The key problem of gene therapy is the choice of an effective vector. Ultrasound-mediated microbubble technique (UMMT) has already shown promising applications in numerous types of tumors apart from cervical carcinoma. In the present study, according to the results of an MTT assay, we initially chose an ultrasound intensity of 0.5 W/cm(2), an ultrasound exposure time of 30 sec and a microbubble concentration of 10% as the optimum experimental condition for wtp53 plasmid transfection into HeLa cells. To further investigate the transfection efficiency of ultrasound combined with microbubbles, RT-PCR analysis was used to examine the mRNA level of p53. The transfection efficiency in the plasmid plus microbubbles and ultrasound group was significantly higher than that of the other groups. Following transfection of the wtp53 gene, flow cytometric analysis showed that the cell cycle of HeLa cells was arrested in the G1 phase. The results of the present study suggest that UMMT, a new gene delivery system, increases the transfection efficiency of the wtp53 gene. Moreover, the growth of HeLa cells was arrested by introducing wtp53. This study may afford a new trend for the gene therapy of cervical carcinoma.
Collapse
|
22
|
Burke CW, Hsiang YHJ, Alexander E, Kilbanov AL, Price RJ. Covalently linking poly(lactic-co-glycolic acid) nanoparticles to microbubbles before intravenous injection improves their ultrasound-targeted delivery to skeletal muscle. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2011; 7:1227-35. [PMID: 21456081 PMCID: PMC3092637 DOI: 10.1002/smll.201001934] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 12/14/2010] [Indexed: 05/26/2023]
Abstract
Intravenously injected nanoparticles can be delivered to skeletal muscle through capillary pores created by the activation of microbubbles with ultrasound; however, strategies that utilize coinjections of free microbubbles and nanoparticles are limited by nanoparticle dilution in the bloodstream. Here, improvement in the delivery of fluorescently labeled ≈150 nm poly(lactic-co-glycolic acid) nanoparticles to skeletal muscle is attempted by covalently linking them to albumin-shelled microbubbles in a composite agent formulation. Studies are performed using an experimental model of peripheral arterial disease, wherein the right and left femoral arteries of BalbC mice are surgically ligated. Four days after arterial ligation, composite agents, coinjected microbubbles and nanoparticles, or nanoparticles alone are administered intravenously and 1 MHz pulsed ultrasound was applied to the left hindlimb. Nanoparticle delivery was assessed at 0, 1, 4, and 24 h post-treatment by fluorescence-mediated tomography. Within the coinjection group, both microbubbles and ultrasound are found to be required for nanoparticle delivery to skeletal muscle. Within the composite agent group, nanoparticle delivery is found to be enhanced 8- to 18-fold over 'no ultrasound' controls, depending on the time of measurement. A maximum of 7.2% of the initial nanoparticle dose per gram of tissue was delivered at 1 hr in the composite agent group, which was significantly greater than in the coinjection group (3.6%). It is concluded that covalently linking 150 nm-diameter poly(lactic-co-glycolic acid) nanoparticles to microbubbles before intravenous injection does improve their delivery to skeletal muscle.
Collapse
Affiliation(s)
- Caitlin W. Burke
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| | - Yu-Han J. Hsiang
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| | - Eben Alexander
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| | - Alexander L. Kilbanov
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
- The Cardiovascular Division, University of Virginia, Charlottesville, Virginia
| | - Richard J. Price
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
- Departments of Radiology & Radiation Oncology, University of Virginia, Charlottesville, VA
| |
Collapse
|
23
|
Deng W, Chen QW, Li XS, Liu H, Niu SQ, Zhou Y, Li GQ, Ke DZ, Mo XG. Bone marrow mesenchymal stromal cells with support of bispecific antibody and ultrasound-mediated microbubbles prevent myocardial fibrosis via the signal transducer and activators of transcription signaling pathway. Cytotherapy 2011; 13:431-40. [DOI: 10.3109/14653249.2010.542458] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
24
|
Chen YC, Jiang LP, Liu NX, Wang ZH, Hong K, Zhang QP. P85, Optison microbubbles and ultrasound cooperate in mediating plasmid DNA transfection in mouse skeletal muscles in vivo. ULTRASONICS SONOCHEMISTRY 2011; 18:513-519. [PMID: 20863738 DOI: 10.1016/j.ultsonch.2010.08.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2010] [Revised: 08/22/2010] [Accepted: 08/31/2010] [Indexed: 05/29/2023]
Abstract
Pluronic block copolymers, a kind of non-ionic surfactant, also known as poloxamers, and ultrasound-targeted microbubble destruction have been respectively investigated as vectors for gene delivery in vitro and in vivo. However, they are limited for clinical application due to the relatively low transfer efficiency of each individual vector. In the present study, we explored if the combination of P85, a pluronic block copolymer, Optison, a microbubble contrast agent and ultrasound enhances the transfection of plasmid DNA in vivo using mouse skeletal muscle models. Plasmid encoding green fluorescent protein (GFP) was respectively conjugated with 0.05%P85, 10%Optison, or 0.05%P85 plus 10%Optison, and injected into mouse tibialis anterior (TA) muscles with or without ultrasound irradiation (1 MHz, 1 W/cm(2), 2 min and 20% duty cycle). Mice were sacrificed 1 week after injection. The TA muscles were collected and cryo-sectioned into a series of 7 μm slices. To assess the efficiency of plasmid DNA transfection, tissue sections were counterstained with DAPI and scored by counting the number of GFP-positive fibers. Meanwhile the area of damaged muscles was measured based on the tissues stained with hematoxylin and eosin. Both P85 and Optison significantly enhanced the delivery of plasmid DNA in mouse TA skeletal muscles (P<0.01 and P<0.05 respectively, compared to saline control). In combination with Ultrasound irradiation, P85 (P<0.01, compared to P85 alone) but not Optison (P>0.05, compared to Optison alone) exerted a more pronounced effect on the transfection efficiency. Furthermore P85-induced gene delivery was higher than that by Optison regardless of the presence of ultrasound (P<0.01). The highest transfection efficiency was observed when P85, Optison and ultrasound irradiation were administrated together (P<0.01, compared to any other treatment in this study). The area of damaged muscles was enlarged by ultrasound irradiation in the presence of Optison microbubbles (P<0.01, compared to those groups without ultrasound irradiation). These results suggest that P85, microbubbles and ultrasound irradiation synergistically enhance plasmid DNA delivery in mouse skeletal muscles in vivo.
Collapse
Affiliation(s)
- Yun-Chao Chen
- Ultrasound Department, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | | | | | | | | | | |
Collapse
|
25
|
Chen YC, Jiang LP, Liu NX, Ding L, Liu XL, Wang ZH, Hong K, Zhang QP. Enhanced Gene Transduction into Skeletal Muscle of Mice In Vivo with Pluronic Block Copolymers and Ultrasound Exposure. Cell Biochem Biophys 2011; 60:267-73. [DOI: 10.1007/s12013-010-9149-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
26
|
Zhou S, Li S, Liu Z, Tang Y, Wang Z, Gong J, Liu C. Ultrasound-targeted microbubble destruction mediated herpes simplex virus-thymidine kinase gene treats hepatoma in mice. J Exp Clin Cancer Res 2010; 29:170. [PMID: 21176239 PMCID: PMC3022677 DOI: 10.1186/1756-9966-29-170] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2010] [Accepted: 12/23/2010] [Indexed: 11/13/2022] Open
Abstract
Objective The purpose of the study was to explore the anti-tumor effect of ultrasound -targeted microbubble destruction mediated herpes simplex virus thymidine kinase (HSV-TK) suicide gene system on mice hepatoma. Methods Forty mice were randomly divided into four groups after the models of subcutaneous transplantation tumors were estabilished: (1) PBS; (2) HSV-TK (3) HSV-TK+ ultrasound (HSV-TK+US); (4) HSV-TK+ultrasound+microbubbles (HSV-TK+US+MB). The TK protein expression in liver cancer was detected by western-blot. Applying TUNEL staining detected tumor cell apoptosis. At last, the inhibition rates and survival time of the animals were compared among all groups. Results The TK protein expression of HSV-TK+MB+US group in tumor-bearing mice tissues were significantly higher than those in other groups. The tumor inhibitory effect of ultrasound-targeted microbubble destruction mediated HSV-TK on mice transplantable tumor was significantly higher than those in other groups (p < 0.05), and can significantly improve the survival time of tumor-bearing mice. Conclusion Ultrasound-targeted microbubble destruction can effectively transfect HSV-TK gene into target tissues and play a significant inhibition effect on tumors, which provides a new strategy for gene therapy in liver cancer.
Collapse
Affiliation(s)
- Shiji Zhou
- Department of Hepatobiliary Surgery, Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing 400010, PR China
| | | | | | | | | | | | | |
Collapse
|
27
|
McKendry JE, Grant CA, Johnson BRG, Coletta PL, Evans JA, Evans SD. Force spectroscopy of streptavidin conjugated lipid coated microbubbles. ACTA ACUST UNITED AC 2010. [DOI: 10.1179/175889610x12865266108541] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
28
|
Kang J, Wu X, Wang Z, Ran H, Xu C, Wu J, Wang Z, Zhang Y. Antitumor effect of docetaxel-loaded lipid microbubbles combined with ultrasound-targeted microbubble activation on VX2 rabbit liver tumors. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2010; 29:61-70. [PMID: 20040776 DOI: 10.7863/jum.2010.29.1.61] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
OBJECTIVE The purpose of the study was to explore the antitumor effect of docetaxel-loaded lipid microbubbles combined with ultrasound-targeted microbubble activation (UTMA) on VX2 rabbit liver tumors. METHODS Docetaxel-loaded lipid microbubbles were made by a mechanical vibration technique. VX2 liver tumor models were established in 90 rabbits, which were randomly divided into 6 groups, including control, docetaxal-loaded lipid microbubbles alone, docetaxal alone, docetaxal combined with ultrasound, pure lipid microbubbles combined with ultrasound, and docetaxel-loaded lipid microbubbles combined with ultrasound (DOC+MB/US). The tumor volume and inhibition rate (IR) of tumor growth were calculated and compared. Apoptosis was detected by terminal deoxyuridine nick end labeling. Proliferating cell nuclear antigen and matrix metalloproteinase 2 (MMP2) protein expression was detected by immunohistochemistry. Caspase 3 and MMP2 messenger RNA (mRNA) expression was detected by in situ hybridization histochemistry. The tumor metastasis rate and survival time of the animals were compared. RESULTS The IR and apoptotic index of the DOC+MB/US group were the highest among all groups, and the proliferating labeling index was the lowest. Matrix metalloproteinase 2 protein and mRNA expression in the DOC+MB/US group was the lowest among all groups, and caspase 3 mRNA expression in the DOC+MB/US group was the highest. The extensive metastasis rate in the DOC+MB/US group was the lowest, and the survival time of the animals in the DOC+MB/US group was the longest. CONCLUSIONS Docetaxel-loaded lipid microbubbles combined with UTMA could inhibit the growth of VX2 rabbit liver tumors by deferring proliferation and promoting apoptosis, which may provide a novel targeted strategy for chemotherapy of liver carcinoma.
Collapse
Affiliation(s)
- Juan Kang
- Department of Gastroenterology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Wells DJ. Electroporation and ultrasound enhanced non-viral gene delivery in vitro and in vivo. Cell Biol Toxicol 2009; 26:21-8. [PMID: 19949971 DOI: 10.1007/s10565-009-9144-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2009] [Accepted: 11/11/2009] [Indexed: 12/18/2022]
Abstract
Non-viral vectors are less efficient than the use of viral vectors for delivery of genetic material to cells in vitro and especially in vivo. However, viral vectors involve the use of foreign proteins that can stimulate both the innate and acquired immune response. In contrast, plasmid DNA can be delivered without carrier proteins and is non-immunogenic. Plasmid gene delivery can be enhanced by the use of physical methods that aid the passage of the plasmid through the cell membrane. Electroporation and microbubble-enhanced ultrasound are two of the most effective physical delivery methods and these can be applied to a range of different cell types in vitro and a broad range of tissues in vivo. Both techniques also have the advantage that, unlike viral vectors, they can be used to target specific tissues with systemic delivery. Although electroporation is often the more efficient of the two, microbubble-enhanced ultrasound causes less damage and is less invasive. This review provides an introduction to the methodology and summarises the range of cells and tissues that have been genetically modified using these techniques.
Collapse
Affiliation(s)
- Dominic J Wells
- Department of Cellular and Molecular Neuroscience, Imperial College London, UK.
| |
Collapse
|
30
|
Gene transfection to retinal ganglion cells mediated by ultrasound microbubbles in vitro. Acad Radiol 2009; 16:1086-94. [PMID: 19541507 DOI: 10.1016/j.acra.2009.03.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2008] [Revised: 03/21/2009] [Accepted: 03/22/2009] [Indexed: 12/22/2022]
Abstract
RATIONALE AND OBJECTIVES To investigate the expression levels of green fluorescence protein (GFP) into retinal ganglion cells (RGCs) in vitro by ultrasound-mediated microbubble destruction (UMMD) and assess the effect of bcl-xl gene on N-methyl-D-aspartate (NMDA)-induced apoptosis in the cultured RGCs by UMMD. MATERIALS AND METHODS pEGFP-N1 was transfected to RGCs in vitro by UMMD and liposome was used as the control. The transfection effect was detected using microscope and flow cytometry qualitatively and quantitatively. Monotetrazolium was adopted to measure the cell vitality. NMDA was used to induce apoptosis in the cultured RGCs, and the bcl-xl gene was transfected into RGCs by UMMD before NMDA-induced apoptosis. The expression of bcl-xl protein in RGCs was assessed by immunohistochemistry assay. The amorphous character of RGCs was revealed by acridine orange and ethidium bromide staining. DNA fragment was detected by agarose gel electrophoresis. RESULTS Ultrasound combined with microbubbles enhanced gene transfection to the cultured cells in some condition. The average transfection rate of pEGFP-N1 with UMMD was 25%. Both ultrasound and microbubble had no effect on cell viability. The expression of bcl-xl protein in transfected and non-transfected RGCs was significantly different. Less apoptotic bodies and no representative DNA fragment were detected in the treatment group. CONCLUSIONS Microbubble destruction can enhance the reporter gene transfection and expression and have a good target. Transfection of bcl-xl gene has an anti-apoptosis effect on the cultured RGCs induced by NMDA with UMMD.
Collapse
|
31
|
Aneja MK, Geiger JP, Himmel A, Rudolph C. Targeted gene delivery to the lung. Expert Opin Drug Deliv 2009; 6:567-83. [DOI: 10.1517/17425240902927841] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
32
|
Xing W, Gang WZ, Yong Z, Yi ZY, Shan XC, Tao RH. Treatment of xenografted ovarian carcinoma using paclitaxel-loaded ultrasound microbubbles. Acad Radiol 2008; 15:1574-9. [PMID: 19000874 DOI: 10.1016/j.acra.2008.06.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2008] [Revised: 06/12/2008] [Accepted: 06/12/2008] [Indexed: 12/11/2022]
Abstract
RATIONALE AND OBJECTIVES The aim of this study was to explore the antitumor effects on mice xenografted ovarian carcinoma using the technique of ultrasound-mediated drug release from paclitaxel-loaded lipid microbubbles (PLMs). MATERIALS AND METHODS Twenty-five ovarian cancer-bearing nude mice were randomly divided into five groups of five mice each. Each group received a unique kind of treatment once a day. These treatments were PLMs combined with ultrasound, intravenous paclitaxel administration, non-drug-loaded microbubbles combined with ultrasound, intravenous PLM administration, and normal saline administration (the control group). After 7 days of consecutive treatment, all mice were sacrificed, and their tumors were harvested to measure volumes and weights. The tumor inhibition rate was calculated by weight. Expressions of vascular endothelial growth factor (VEGF) and p53 in tumor tissues were detected by immunohistochemical staining. RESULTS Mean tumor volume and weight were the lowest in the first group (PLMs combined with ultrasound), so this group's tumor inhibition rate was the highest (P < .05). On immunohistology, VEGF and p53 expression levels were lowest (P < .05) in the first group. CONCLUSION Ultrasound irradiation mediates PLM destruction so that the drug is released from the vehicles at the same time. It helps achieve targeted chemotherapy in tumor tissues. This technique has potential to be adopted as a novel tool for ovarian cancer chemotherapy.
Collapse
|
33
|
Abstract
Ultrasound exposure (USE) in the presence of microbubbles (MCB) (e.g. contrast agents used to enhance ultrasound imaging) increases plasmid transfection efficiency in vitro by several orders of magnitude. Formation of short-lived pores in the plasma membrane ('sonoporation'), up to 100 nm in effective diameter lasting a few seconds, is implicated as the dominant mechanism, associated with acoustic cavitation. Ultrasound enhanced gene transfer (UEGT) has also been successfully achieved in vivo, with reports of spatially restricted and therapeutically relevant levels of transgene expression. Loading MCB with nucleic acids and/or disease-targeting ligands may further improve the efficiency and specificity of UEGT such that clinical testing becomes a realistic prospect.
Collapse
Affiliation(s)
- C M H Newman
- Cardiovascular Research Unit, School of Medicine and Biomedical Sciences, University of Sheffield, Sheffield, UK.
| | | |
Collapse
|