1
|
Xin J, Lu X, Cao J, Wu W, Liu Q, Wang D, Zhou X, Ding D. Fluorinated Organic Polymers for Cancer Drug Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2404645. [PMID: 38678386 DOI: 10.1002/adma.202404645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 04/22/2024] [Indexed: 04/30/2024]
Abstract
In the realm of cancer therapy, the spotlight is on nanoscale pharmaceutical delivery systems, especially polymer-based nanoparticles, for their enhanced drug dissolution, extended presence in the bloodstream, and precision targeting achieved via surface engineering. Leveraging the amplified permeation and retention phenomenon, these systems concentrate therapeutic agents within tumor tissues. Nonetheless, the hurdles of systemic toxicity, biological barriers, and compatibility with living systems persist. Fluorinated polymers, distinguished by their chemical idiosyncrasies, are poised for extensive biomedical applications, notably in stabilizing drug metabolism, augmenting lipophilicity, and optimizing bioavailability. Material science heralds the advent of fluorinated polymers that, by integrating fluorine atoms, unveil a suite of drug delivery merits: the hydrophobic traits of fluorinated alkyl chains ward off lipid or protein disruption, the carbon-fluorine bond's stability extends the drug's lifecycle in the system, and a lower alkalinity coupled with a diminished ionic charge bolsters the drug's ability to traverse cellular membranes. This comprehensive review delves into the utilization of fluorinated polymers for oncological pharmacotherapy, elucidating their molecular architecture, synthetic pathways, and functional attributes, alongside an exploration of their empirical strengths and the quandaries they encounter in both experimental and clinical settings.
Collapse
Affiliation(s)
- Jingrui Xin
- College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Xue Lu
- Frontiers Science Center for New Organic Matter, Nankai International Advanced Research Institute (Shenzhen, Futian), and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Jimin Cao
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, and First Clinical Medical College, Shanxi Medical University, Taiyuan, 030001, China
| | - Weihui Wu
- Frontiers Science Center for New Organic Matter, Nankai International Advanced Research Institute (Shenzhen, Futian), and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Qian Liu
- Department of Urology, Tianjin First Central Hospital, Tianjin, 300192, China
| | - Deping Wang
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, and First Clinical Medical College, Shanxi Medical University, Taiyuan, 030001, China
| | - Xin Zhou
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, and First Clinical Medical College, Shanxi Medical University, Taiyuan, 030001, China
| | - Dan Ding
- Frontiers Science Center for New Organic Matter, Nankai International Advanced Research Institute (Shenzhen, Futian), and College of Life Sciences, Nankai University, Tianjin, 300071, China
| |
Collapse
|
2
|
Ren H, Xiang S, Liu A, Wang Q, Zhou N, Hu Z. A noval noninvasive targeted therapy for osteosarcoma: the combination of LIFU and ultrasound-magnetic-mediated SPIO/TP53/PLGA nanobubble. Front Bioeng Biotechnol 2024; 12:1418903. [PMID: 39007051 PMCID: PMC11239426 DOI: 10.3389/fbioe.2024.1418903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 06/10/2024] [Indexed: 07/16/2024] Open
Abstract
Purpose Osteosarcoma (OS) is the most common type of primary malignant bone tumor. Transducing a functional TP53 gene can effectively inhibit OS cell activity. Poly lactic acid-glycolic acid (PLGA) nanobubbles (NBs) mediated by focused ultrasound (US) can introduce exogenous genes into target cells in animal models, but this technique relies on the passive free diffusion of agents across the body. The inclusion of superparamagnetic iron oxide (SPIO) in microbubbles allows for magnetic-based tissue localization. A low-intensity-focused ultrasound (LIFU) instrument was developed at our institute, and different intensities of LIFU can either disrupt the NBs (RLI-LIFU) or exert cytocidal effects on the target tissues (RHI-LIFU). Based on these data, we performed US-magnetic-mediated TP53-NB destruction and investigated its ability to inhibit OS growth when combined with LIFU both in vitro and in vivo. Methods Several SPIO/TP53/PLGA (STP) NB variants were prepared and characterized. For the in vitro experiments, HOS and MG63 cells were randomly assigned into five treatment groups. Cell proliferation and the expression of TP53 were detected by CCK8, qRT-PCR and Western blotting, respectively. In vivo, tumor-bearing nude mice were randomly assigned into seven treatment groups. The iron distribution of Perls' Prussian blue-stained tissue sections was determined by optical microscopy. TUNEL-DAPI was performed to examine apoptosis. TP53 expression was detected by qRT-PCR and immunohistochemistry. Results SPIO/TP53/PLGA NBs with a particle size of approximately 200 nm were prepared successfully. For in vitro experiments, ultrasound-targeted transfection of TP53 overexpression in OS cells and efficient inhibition of OS proliferation have been demonstrated. Furthermore, in a tumor-bearing nude mouse model, RLI-LIFU-magnetic-mediated SPIO/TP53/PLGA NBs increased the transfection efficiency of the TP53 plasmid, resulting in apoptosis. Adding RHI-LIFU to the treatment regimen significantly increased the apoptosis of OS cells in vivo. Conclusion Combining LIFU and US-magnetic-mediated SPIO/TP53/PLGA NB destruction is potentially a novel noninvasive and targeted therapy for OS.
Collapse
Affiliation(s)
- Honglei Ren
- Department of Orthopedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Orthopedic Surgery, ChongQing Red Cross Hospital (People's Hospital of JiangBei District), Chongqing, China
| | - Shanlin Xiang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Aiguo Liu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Henan University, Kaifeng, China
| | - Qian Wang
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Nian Zhou
- Department of Orthopedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhenming Hu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Orthopedic Surgery, The University-Town Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
3
|
Yang CD, Jessen J, Lin KY. Ultrasound-assisted ocular drug delivery: A review of current evidence. JOURNAL OF CLINICAL ULTRASOUND : JCU 2022; 50:685-693. [PMID: 35474512 DOI: 10.1002/jcu.23214] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/11/2022] [Accepted: 04/15/2022] [Indexed: 06/14/2023]
Abstract
Efficient ocular drug delivery is a challenging clinical problem with various therapeutic options but no clearly preferred methodology. Given the ubiquity of ultrasound as a diagnostic technique, the safety profile of ultrasound in an ocular context, and the prospect of custom-made ultrasound-sensitive contrast agents, ultrasound presents an attractive ocular drug delivery modality. In this review, we evaluate our present understanding of ultrasound as it relates to ocular drug delivery and significant knowledge gaps in the field. In doing so, we hope to call attention to a potentially novel drug delivery pathway that could be manipulated to treat or cure ocular diseases.
Collapse
Affiliation(s)
- Christopher D Yang
- Department of Ophthalmology, University of California, Irvine School of Medicine, Irvine, California, USA
- Gavin Herbert Eye Institute, University of California, Irvine, Irvine, California, USA
| | - Jordan Jessen
- Gavin Herbert Eye Institute, University of California, Irvine, Irvine, California, USA
| | - Ken Y Lin
- Department of Ophthalmology, University of California, Irvine School of Medicine, Irvine, California, USA
- Gavin Herbert Eye Institute, University of California, Irvine, Irvine, California, USA
- Department of Biomedical Engineering, University of California, Irvine, California, USA
| |
Collapse
|
4
|
Applications of Ultrasound-Mediated Gene Delivery in Regenerative Medicine. Bioengineering (Basel) 2022; 9:bioengineering9050190. [PMID: 35621468 PMCID: PMC9137703 DOI: 10.3390/bioengineering9050190] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/22/2022] [Accepted: 04/23/2022] [Indexed: 11/21/2022] Open
Abstract
Research on the capability of non-viral gene delivery systems to induce tissue regeneration is a continued effort as the current use of viral vectors can present with significant limitations. Despite initially showing lower gene transfection and gene expression efficiencies, non-viral delivery methods continue to be optimized to match that of their viral counterparts. Ultrasound-mediated gene transfer, referred to as sonoporation, occurs by the induction of transient membrane permeabilization and has been found to significantly increase the uptake and expression of DNA in cells across many organ systems. In addition, it offers a more favorable safety profile compared to other non-viral delivery methods. Studies have shown that microbubble-enhanced sonoporation can elicit significant tissue regeneration in both ectopic and disease models, including bone and vascular tissue regeneration. Despite this, no clinical trials on the use of sonoporation for tissue regeneration have been conducted, although current clinical trials using sonoporation for other indications suggest that the method is safe for use in the clinical setting. In this review, we describe the pre-clinical studies conducted thus far on the use of sonoporation for tissue regeneration. Further, the various techniques used to increase the effectiveness and duration of sonoporation-induced gene transfer, as well as the obstacles that may be currently hindering clinical translation, are explored.
Collapse
|
5
|
Singh R, Yang X. A 3D finite element model to study the cavitation induced stresses on blood-vessel wall during the ultrasound-only phase of photo-mediated ultrasound therapy. AIP ADVANCES 2022; 12:045020. [PMID: 35465057 PMCID: PMC9020880 DOI: 10.1063/5.0082429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/28/2022] [Indexed: 06/14/2023]
Abstract
Photo-mediated ultrasound therapy (PUT) is a novel technique utilizing synchronized ultrasound and laser to generate enhanced cavitation inside blood vessels. The enhanced cavitation inside blood vessels induces bio-effects, which can result in the removal of micro-vessels and the reduction in local blood perfusion. These bio-effects have the potential to treat neovascularization diseases in the eye, such as age-related macular degeneration and diabetic retinopathy. Currently, PUT is in the preclinical stage, and various PUT studies on in vivo rabbit eye models have shown successful removal of micro-vessels. PUT is completely non-invasive and particle-free as opposed to current clinical treatments such as anti-vascular endothelial growth factor therapy and photodynamic therapy, and it precisely removes micro-vessels without damaging the surrounding tissue, unlike laser photocoagulation therapy. The stresses produced by oscillating bubbles during PUT are responsible for the induced bio-effects in blood vessels. In our previous work, stresses induced during the first phase of PUT due to combined ultrasound and laser irradiation were studied using a 2D model. In this work, stresses induced during the third or last phase of PUT due to ultrasound alone were studied using a 3D finite element method-based numerical model. The results showed that the circumferential and shear stress increased as the bubble moves from the center of the vessel toward the vessel wall with more than a 16 times increase in shear stress from 1.848 to 31.060 kPa as compared to only a 4 times increase in circumferential stress from 211 to 906 kPa for a 2 µm bubble placed inside a 10 µm vessel on the application of 1 MHz ultrasound frequency and 130 kPa amplitude. In addition, the stresses decreased as the bubble was placed in smaller sized vessels with a larger decrease in circumferential stress. The changes in shear stress were found to be more dependent on the bubble-vessel wall distance, and the changes in circumferential stress were more dependent on the bubble oscillation amplitude. Moreover, the bubble shape changed to an ellipsoidal with a higher oscillation amplitude in the vessel's axial direction as it was moved closer to the vessel wall, and the bubble oscillation amplitude decreased drastically as it was placed in vessels of a smaller size.
Collapse
Affiliation(s)
| | - Xinmai Yang
- Author to whom correspondence should be addressed:
| |
Collapse
|
6
|
Walsh AP, Gordon HN, Peter K, Wang X. Ultrasonic particles: An approach for targeted gene delivery. Adv Drug Deliv Rev 2021; 179:113998. [PMID: 34662671 PMCID: PMC8518240 DOI: 10.1016/j.addr.2021.113998] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/24/2021] [Accepted: 10/05/2021] [Indexed: 02/07/2023]
Abstract
Gene therapy has been widely investigated for the treatment of genetic, acquired, and infectious diseases. Pioneering work utilized viral vectors; however, these are suspected of causing serious adverse events, resulting in the termination of several clinical trials. Non-viral vectors, such as lipid nanoparticles, have attracted significant interest, mainly due to their successful use in vaccines in the current COVID-19 pandemic. Although they allow safe delivery, they come with the disadvantage of off-target delivery. The application of ultrasound to ultrasound-sensitive particles allows for a direct, site-specific transfer of genetic materials into the organ/site of interest. This process, termed ultrasound-targeted gene delivery (UTGD), also increases cell membrane permeability and enhances gene uptake. This review focuses on the advances in ultrasound and the development of ultrasonic particles for UTGD across a range of diseases. Furthermore, we discuss the limitations and future perspectives of UTGD.
Collapse
Affiliation(s)
- Aidan P.G. Walsh
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Department of Medicine, Monash University, Melbourne, VIC, Australia
| | - Henry N. Gordon
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Department of Biochemistry and Pharmacology, University of Melbourne, VIC, Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Department of Medicine, Monash University, Melbourne, VIC, Australia,Department of Cardiometabolic Health, University of Melbourne, VIC, Australia,La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Xiaowei Wang
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Department of Medicine, Monash University, Melbourne, VIC, Australia,Department of Cardiometabolic Health, University of Melbourne, VIC, Australia,La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia,Corresponding author at: Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| |
Collapse
|
7
|
Wang X, Wang D, Xia P, Cheng K, Wang Q, Wang X, Lin Q, Song J, Chen A, Li X. Ultrasound-targeted simvastatin-loaded microbubble destruction promotes OA cartilage repair by modulating the cholesterol efflux pathway mediated by PPARγ in rabbits. Bone Joint Res 2021; 10:693-703. [PMID: 34666502 PMCID: PMC8559971 DOI: 10.1302/2046-3758.1010.bjr-2021-0162.r3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Aims To evaluate the effect of ultrasound-targeted simvastatin-loaded microbubble destruction (UTMDSV) for alleviation of the progression of osteoarthritis (OA) in rabbits through modulation of the peroxisome proliferator-activated receptor (PPARγ). Methods In vitro, OA chondrocytes were treated with ultrasound (US), US-targeted microbubble destruction (UTMD), simvastatin (SV), and UTMDSV on alternate days for four weeks. Chondrocytes were also treated with PPARγ inhibitor, PPARγ inhibitor+ UTMDSV, and UTMDSV. The cholesterol efflux rate and triglyceride levels were measured using an assay kit and oil red O staining, respectively. In vivo, the OA rabbits were treated with a single intra-articular injection of UTMD, SV, and UTMDSV every seven days for four weeks. Cartilage histopathology was assessed by safranin-O staining and the Mankin score. Total cholesterol (TC) and high-density lipoprotein-cholesterol (HDL-C) in rabbit knee synovial fluid were detected by enzyme-marker assay. Aggrecan, collagen II, and PPARγ expression levels were analyzed by Western blotting (WB). Results In vitro, UTMDSV significantly increased the cholesterol efflux rate and aggrecan, collagen II, and PPARγ levels in OA chondrocytes; these effects were blocked by the PPARγ inhibitor. In vivo, UTMDSV significantly increased aggrecan, collagen II, PPARγ, and HDL-C levels, while TC levels and Mankin scores were decreased compared with the UTMD, SV, OA, and control groups. Conclusion UTMDSV promotes cartilage extracellular matrix synthesis by modulating the PPARγ-mediated cholesterol efflux pathway in OA rabbits. Cite this article: Bone Joint Res 2021;10(10):693–703.
Collapse
Affiliation(s)
- Xinwei Wang
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Danbi Wang
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Peng Xia
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Kai Cheng
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Qi Wang
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiaoju Wang
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Qiang Lin
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jiulong Song
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Anliang Chen
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xueping Li
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
8
|
Mahfouz M, Maruyama R, Yokota T. Inotersen for the Treatment of Hereditary Transthyretin Amyloidosis. Methods Mol Biol 2021; 2176:87-98. [PMID: 32865784 DOI: 10.1007/978-1-0716-0771-8_6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Hereditary transthyretin amyloidosis (hATTR) is a rare autosomal dominant condition in which mutations in the transthyretin gene cause amyloid fibrils to develop and deposit into tissues, affecting primarily the nerves and heart causing polyneuropathy and cardiomyopathy respectively. Standard treatment has been liver transplants to try and eliminate the mutated transthyretin products as the liver is the main source of transthyretin production. A new drug named inotersen (brand name Tagsedi), also known as IONIS-TTRRX, has been approved by the United States Food and Drug Agency, Health Canada, and European Commission in 2018, and introduced to the market for patients in stage 1 and stage 2 hATTR polyneuropathy. Inotersen is a second-generation antisense oligonucleotide with 2'-O-methoxyethyl modification designed to bind to the 3' untranslated region of the transthyretin mRNA in the nucleus of the liver cells. By doing so, it prevents the production of the mutant and wild-type forms of transthyretin, impeding the progression of the disease. In this article, the mechanism of action and safety profile of inotersen will be discussed along with some future directions following its approval.
Collapse
Affiliation(s)
- Maria Mahfouz
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- School of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Rika Maruyama
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Toshifumi Yokota
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
- The Friends of Garrett Cumming Research & Muscular Dystrophy Canada HM Toupin Neurological Science Research Chair, Edmonton, AB, Canada.
| |
Collapse
|
9
|
Hosseinpour S, Walsh LJ. Laser-assisted nucleic acid delivery: A systematic review. JOURNAL OF BIOPHOTONICS 2021; 14:e202000295. [PMID: 32931155 DOI: 10.1002/jbio.202000295] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 08/26/2020] [Accepted: 09/13/2020] [Indexed: 06/11/2023]
Abstract
Gene therapy has become an effective treatment modality for some conditions. Laser light may augment or enhance gene therapy through photomechanical, photothermal, and photochemical. This review examined the evidence base for laser therapy to enhance nucleic acid transfection in mammalian cells. An electronic search of MEDLINE, Scopus, EMBASE, Web of Science, and Google Scholar was performed, covering all available years. The preferred reporting items for systematic reviews and meta-analyses guideline for systematic reviews was used for designing the study and analyzing the results. In total, 49 studies of laser irradiation for nucleic acid delivery were included. Key approaches were optoporation, photomechanical gene transfection, and photochemical internalization. Optoporation is better suited to cells in culture, photomechanical and photochemical approaches appear well suited to in vivo use. Additional studies explored the impact of photothermal for enhancing gene transfection. Each approach has merits and limitations. Augmenting nucleic acid delivery using laser irradiation is a promising method for improving gene therapy. Laser protocols can be non-invasive because of the penetration of desirable wavelengths of light, but it depends on various parameters such as power density, treatment duration, irradiation mode, etc. The current protocols show low efficiency, and there is a need for further work to optimize irradiation parameters.
Collapse
Affiliation(s)
- Sepanta Hosseinpour
- School of Dentistry, Oral Health Centre, The University of Queensland, Brisbane, Australia
| | - Laurence J Walsh
- School of Dentistry, Oral Health Centre, The University of Queensland, Brisbane, Australia
| |
Collapse
|
10
|
Kim D, Lee JH, Moon H, Seo M, Han H, Yoo H, Seo H, Lee J, Hong S, Kim P, Lee HJ, Chung JW, Kim H. Development and evaluation of an ultrasound-triggered microbubble combined transarterial chemoembolization (TACE) formulation on rabbit VX2 liver cancer model. Am J Cancer Res 2021; 11:79-92. [PMID: 33391462 PMCID: PMC7681087 DOI: 10.7150/thno.45348] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 09/17/2020] [Indexed: 02/06/2023] Open
Abstract
Transarterial chemoembolization (TACE) is an image-guided locoregional therapy used for the treatment of patients with primary or secondary liver cancer. However, conventional TACE formulations are rapidly dissociated due to the instability of the emulsion, resulting in insufficient local drug concentrations in the target tumor. Methods: To overcome these limitations, a doxorubicin-loaded albumin nanoparticle-conjugated microbubble complex in an iodized oil emulsion (DOX-NPs-MB complex in Lipiodol) has been developed as a new ultrasound-triggered TACE formulation. Results: (1) Microbubbles enhanced therapeutic efficacy by effectively delivering doxorubicin- loaded nanoparticles into liver tumors via sonoporation under ultrasound irradiation (US+). (2) Microbubbles constituting the complex retained their function as an ultrasound contrast agent in Lipiodol. In a rabbit VX2 liver cancer model, the in vivo study of DOX-NPs-MB complex in Lipiodol (US+) decreased the viability of tumor more than the conventional TACE formulation, and in particular, effectively killed cancer cells in the tumor periphery. Conclusion: Incorporation of doxorubicin-loaded microbubble in the TACE formulation facilitated drug delivery to the tumor with real-time monitoring and enhanced the therapeutic efficacy of TACE. Thus, the enhanced TACE formulation may represent a new treatment strategy against liver cancer.
Collapse
|
11
|
Decker RE, Lamantia ZE, Emrick TS, Figueiredo ML. Sonodelivery in Skeletal Muscle: Current Approaches and Future Potential. Bioengineering (Basel) 2020; 7:E107. [PMID: 32916815 PMCID: PMC7552685 DOI: 10.3390/bioengineering7030107] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/27/2020] [Accepted: 09/04/2020] [Indexed: 11/16/2022] Open
Abstract
There are currently multiple approaches to facilitate gene therapy via intramuscular gene delivery, such as electroporation, viral delivery, or direct DNA injection with or without polymeric carriers. Each of these methods has benefits, but each method also has shortcomings preventing it from being established as the ideal technique. A promising method, ultrasound-mediated gene delivery (or sonodelivery) is inexpensive, widely available, reusable, minimally invasive, and safe. Hurdles to utilizing sonodelivery include choosing from a large variety of conditions, which are often dependent on the equipment and/or research group, and moderate transfection efficiencies when compared to some other gene delivery methods. In this review, we provide a comprehensive look at the breadth of sonodelivery techniques for intramuscular gene delivery and suggest future directions for this continuously evolving field.
Collapse
Affiliation(s)
- Richard E. Decker
- Department of Basic Medical Sciences, Purdue University, 625 Harrison St., West Lafayette, IN 47907, USA; (R.E.D.); (Z.E.L.)
| | - Zachary E. Lamantia
- Department of Basic Medical Sciences, Purdue University, 625 Harrison St., West Lafayette, IN 47907, USA; (R.E.D.); (Z.E.L.)
| | - Todd S. Emrick
- Department of Polymer Science & Engineering, University of Massachusetts, 120 Governors Drive, Amherst, MA 01003, USA;
| | - Marxa L. Figueiredo
- Department of Basic Medical Sciences, Purdue University, 625 Harrison St., West Lafayette, IN 47907, USA; (R.E.D.); (Z.E.L.)
| |
Collapse
|
12
|
Oliva N, Almquist BD. Spatiotemporal delivery of bioactive molecules for wound healing using stimuli-responsive biomaterials. Adv Drug Deliv Rev 2020; 161-162:22-41. [PMID: 32745497 DOI: 10.1016/j.addr.2020.07.021] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/03/2020] [Accepted: 07/23/2020] [Indexed: 12/28/2022]
Abstract
Wound repair is a fascinatingly complex process, with overlapping events in both space and time needed to pave a pathway to successful healing. This additional complexity presents challenges when developing methods for the controlled delivery of therapeutics for wound repair and tissue engineering. Unlike more traditional applications, where biomaterial-based depots increase drug solubility and stability in vivo, enhance circulation times, and improve retention in the target tissue, when aiming to modulate wound healing, there is a desire to enable localised, spatiotemporal control of multiple therapeutics. Furthermore, many therapeutics of interest in the context of wound repair are sensitive biologics (e.g. growth factors), which present unique challenges when designing biomaterial-based delivery systems. Here, we review the diverse approaches taken by the biomaterials community for creating stimuli-responsive materials that are beginning to enable spatiotemporal control over the delivery of therapeutics for applications in tissue engineering and regenerative medicine.
Collapse
|
13
|
Karki A, Giddings E, Carreras A, Champagne D, Fortner K, Rincon M, Wu J. Sonoporation as an Approach for siRNA delivery into T cells. ULTRASOUND IN MEDICINE & BIOLOGY 2019; 45:3222-3231. [PMID: 31540758 DOI: 10.1016/j.ultrasmedbio.2019.06.406] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 05/29/2019] [Accepted: 06/07/2019] [Indexed: 06/10/2023]
Abstract
Delivery of small interfering RNAs (siRNAs) into primary T cells is quite challenging because they are non-proliferating cells and are difficult to transfect with non-viral approaches. Because sonoporation is independent of the proliferation status of cells and siRNA acts in the cell cytoplasm, we investigated whether sonoporation could be used to deliver siRNA into mouse and human T cells. Cells mixed with Definity microbubbles and siRNA were sonicated with a non-focused transducer of center frequency 2.20 MHz producing ultrasound at a 10% duty cycle, pulse repetition frequency of 2.20 kHz and spatial average temporal average ultrasound intensity of 1.29 W/cm2 for 5 s and then examined for siRNA fluorescence by flow cytometry analysis. These sonoporation conditions resulted in high-efficiency transfection of siRNA in mouse and human T cells. Further, the efficacy of siRNA delivery by sonoporation was illustrated by the successful visualization of decreased methylation-controlled J protein expression in mouse and human CD8 T cells via Western blot analysis. The results provide the first evidence that sonoporation is a novel approach to delivery of siRNA into fresh isolated mouse and human T cells in vitro, and might be used for in vivo studies in the future.
Collapse
Affiliation(s)
- Alina Karki
- Department of Physics, University of Vermont, Burlington, Vermont, USA
| | - Emily Giddings
- Division of Immunobiology, University of Vermont, Burlington, Vermont, USA
| | - Ana Carreras
- Division of Immunobiology, University of Vermont, Burlington, Vermont, USA
| | - Devin Champagne
- Division of Immunobiology, University of Vermont, Burlington, Vermont, USA
| | - Karen Fortner
- Division of Immunobiology, University of Vermont, Burlington, Vermont, USA
| | - Mercedes Rincon
- Division of Immunobiology, University of Vermont, Burlington, Vermont, USA
| | - Junru Wu
- Department of Physics, University of Vermont, Burlington, Vermont, USA.
| |
Collapse
|
14
|
He Y, Liu Y, Sun Z, Han F, Tang JZ, Gao R, Wang G. The proper strategy to compress and protect plasmid DNA in the Pluronic L64-electropulse system for enhanced intramuscular gene delivery. Regen Biomater 2019; 6:289-298. [PMID: 31616566 PMCID: PMC6783702 DOI: 10.1093/rb/rby028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 10/30/2018] [Accepted: 11/26/2018] [Indexed: 12/14/2022] Open
Abstract
Intramuscular expression of functional proteins is a promising strategy for therapeutic purposes. Previously, we developed an intramuscular gene delivery method by combining Pluronic L64 and optimized electropulse, which is among the most efficient methods to date. However, plasmid DNAs (pDNAs) in this method were not compressed, making them unstable and inefficient in vivo. We considered that a proper compression of pDNAs by an appropriate material should facilitate gene expression in this L64-electropulse system. Here, we reported our finding of such a material, Epigallocatechin gallate (EGCG), a natural compound in green teas, which could compress and protect pDNAs and significantly increase intramuscular gene expression in the L64-electropulse system. Meanwhile, we found that polyethylenimine (PEI) could also slightly improve exogenous gene expression in the optimal procedure. By analysing the characteristic differences between EGCG and PEI, we concluded that negatively charged materials with strong affinity to nucleic acids and/or other properties suitable for gene delivery, such as EGCG, are better alternatives than cationic materials (like PEI) for muscle-based gene delivery. The results revealed that a critical principle for material/pDNA complex benefitting intramuscular gene delivery/expression is to keep the complex negatively charged. This proof-of-concept study displays the breakthrough in compressing pDNAs and provides a principle and strategy to develop more efficient intramuscular gene delivery systems for therapeutic applications.
Collapse
Affiliation(s)
- Yutong He
- National Engineering Research Center for Biomaterials
| | - Yili Liu
- National Engineering Research Center for Biomaterials
- Key Laboratory for Bio-Resource and Eco-Environment of Ministry Education, College of Life Science, Sichuan University, No. 29, Wangjiang Road, Chengdu, Sichuan, P.R. China
| | - Zhe Sun
- National Engineering Research Center for Biomaterials
| | - Fei Han
- National Engineering Research Center for Biomaterials
| | - James Zhenggui Tang
- Research Institute in Healthcare Science, Faculty of Science and Engineering, School of Pharmacy, University of Wolverhampton, Wolverhampton, UK
| | - Rong Gao
- Key Laboratory for Bio-Resource and Eco-Environment of Ministry Education, College of Life Science, Sichuan University, No. 29, Wangjiang Road, Chengdu, Sichuan, P.R. China
| | - Gang Wang
- National Engineering Research Center for Biomaterials
| |
Collapse
|
15
|
Synergistic anti-tumor effect of paclitaxel and miR-34a combined with ultrasound microbubbles on cervical cancer in vivo and in vitro. Clin Transl Oncol 2019; 22:60-69. [DOI: 10.1007/s12094-019-02131-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 04/08/2019] [Indexed: 01/01/2023]
|
16
|
Nonviral ultrasound-mediated gene delivery in small and large animal models. Nat Protoc 2019; 14:1015-1026. [PMID: 30804568 DOI: 10.1038/s41596-019-0125-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 12/18/2018] [Indexed: 12/15/2022]
Abstract
Ultrasound-mediated gene delivery (sonoporation) is a minimally invasive, nonviral and clinically translatable method of gene therapy. This method offers a favorable safety profile over that of viral vectors and is less invasive as compared with other physical gene delivery approaches (e.g., electroporation). We have previously used sonoporation to overexpress transgenes in different skeletal tissues in order to induce tissue regeneration. Here, we provide a protocol that could easily be adapted to address various other targets of tissue regeneration or additional applications, such as cancer and neurodegenerative diseases. This protocol describes how to prepare, conduct and optimize ultrasound-mediated gene delivery in both a murine and a porcine animal model. The protocol includes the preparation of a microbubble-DNA mix and in vivo sonoporation under ultrasound imaging. Ultrasound-mediated gene delivery can be accomplished within 10 min. After DNA delivery, animals can be followed to monitor gene expression, protein secretion and other transgene-specific outcomes, including tissue regeneration. This procedure can be accomplished by a competent graduate student or technician with prior experience in ultrasound imaging or in performing in vivo procedures.
Collapse
|
17
|
Do HD, Couillaud BM, Doan BT, Corvis Y, Mignet N. Advances on non-invasive physically triggered nucleic acid delivery from nanocarriers. Adv Drug Deliv Rev 2019; 138:3-17. [PMID: 30321618 DOI: 10.1016/j.addr.2018.10.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 09/14/2018] [Accepted: 10/08/2018] [Indexed: 12/15/2022]
Abstract
Nucleic acids (NAs) have been considered as promising therapeutic agents for various types of diseases. However, their clinical applications still face many limitations due to their charge, high molecular weight, instability in biological environment and low levels of transfection. To overcome these drawbacks, therapeutic NAs should be carried in a stable nanocarrier, which can be viral or non-viral vectors, and released at specific target site. Various controllable gene release strategies are currently being evaluated with interesting results. Endogenous stimuli-responsive systems, for example pH-, redox reaction-, enzymatic-triggered approaches have been widely studied based on the physiological differences between pathological and normal tissues. Meanwhile, exogenous triggered release strategies require the use of externally non-invasive physical triggering signals such as light, heat, magnetic field and ultrasound. Compared to internal triggered strategies, external triggered gene release is time and site specifically controllable through active management of outside stimuli. The signal induces changes in the stability of the delivery system or some specific reactions which lead to endosomal escape and/or gene release. In the present review, the mechanisms and examples of exogenous triggered gene release approaches are detailed. Challenges and perspectives of such gene delivery systems are also discussed.
Collapse
|
18
|
Du X, Wang J, Zhou Q, Zhang L, Wang S, Zhang Z, Yao C. Advanced physical techniques for gene delivery based on membrane perforation. Drug Deliv 2018; 25:1516-1525. [PMID: 29968512 PMCID: PMC6058615 DOI: 10.1080/10717544.2018.1480674] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Gene delivery as a promising and valid tool has been used for treating many serious diseases that conventional drug therapies cannot cure. Due to the advancement of physical technology and nanotechnology, advanced physical gene delivery methods such as electroporation, magnetoporation, sonoporation and optoporation have been extensively developed and are receiving increasing attention, which have the advantages of briefness and nontoxicity. This review introduces the technique detail of membrane perforation, with a brief discussion for future development, with special emphasis on nanoparticles mediated optoporation that have developed as an new alternative transfection technique in the last two decades. In particular, the advanced physical approaches development and new technology are highlighted, which intends to stimulate rapid advancement of perforation techniques, develop new delivery strategies and accelerate application of these techniques in clinic.
Collapse
Affiliation(s)
- Xiaofan Du
- a Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Analytical Technology and Instrumentation , School of Life Science and Technology, Xi'an Jiaotong University , Xi'an , People's Republic of China
| | - Jing Wang
- a Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Analytical Technology and Instrumentation , School of Life Science and Technology, Xi'an Jiaotong University , Xi'an , People's Republic of China
| | - Quan Zhou
- a Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Analytical Technology and Instrumentation , School of Life Science and Technology, Xi'an Jiaotong University , Xi'an , People's Republic of China
| | - Luwei Zhang
- a Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Analytical Technology and Instrumentation , School of Life Science and Technology, Xi'an Jiaotong University , Xi'an , People's Republic of China
| | - Sijia Wang
- a Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Analytical Technology and Instrumentation , School of Life Science and Technology, Xi'an Jiaotong University , Xi'an , People's Republic of China
| | - Zhenxi Zhang
- a Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Analytical Technology and Instrumentation , School of Life Science and Technology, Xi'an Jiaotong University , Xi'an , People's Republic of China
| | - Cuiping Yao
- a Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Analytical Technology and Instrumentation , School of Life Science and Technology, Xi'an Jiaotong University , Xi'an , People's Republic of China
| |
Collapse
|
19
|
Ultrasound triggered phase-change nanodroplets for doxorubicin prodrug delivery and ultrasound diagnosis: An in vitro study. Colloids Surf B Biointerfaces 2018; 174:416-425. [PMID: 30481702 DOI: 10.1016/j.colsurfb.2018.11.046] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 10/24/2018] [Accepted: 11/19/2018] [Indexed: 11/22/2022]
Abstract
Ultrasound-triggered delivery system is among the various multifunctional and stimuli-responsive strategies that hold great potential as a robust solution to the challenges of localized drug delivery and gene therapy. In this work, we developed an ultrasound-triggered delivery system PFP/C9F17-PAsp(DET)/CAD/PGA-g-mPEG nanodroplet, which combined ultrasound responsive phase-change contrast agent, acid-cleavable doxorubicin prodrug and cationic amphiphilic fluorinated polymer carrier, aiming to achieve both high imaging contrast and preferable ultrasound-triggered anti-cancer therapeutic effect. The optimized nanodroplets were characterized as monodispersed particles with a diameter of about 400 nm, slightly positive surface charge and high drug-loading efficiency. The functional augmenter PGA-g-mPEG provided the nanodroplets good sustainability, low cytotoxicity and good serum compatibility, as confirmed by stability and biocompatibility tests. In ultrasound imaging study, the nanodroplets produced significant contrast with ultrasound irradiation (3.5 MHz, MI = 0.08) at 37 ℃. Cell uptake and cytotoxicity studies in HepG2 and CT-26 cells showed the enhanced drug uptake and therapeutic effect with the combination of nanodroplets and ultrasound irradiation. These results suggest that the PFP/CAD-loaded phase change nano-emulsion can be utilized as an efficient theranostic agent for both ultrasound contrast imaging and drug delivery.
Collapse
|
20
|
Bez M, Kremen TJ, Tawackoli W, Avalos P, Sheyn D, Shapiro G, Giaconi JC, Ben David S, Snedeker JG, Gazit Z, Ferrara KW, Gazit D, Pelled G. Ultrasound-Mediated Gene Delivery Enhances Tendon Allograft Integration in Mini-Pig Ligament Reconstruction. Mol Ther 2018; 26:1746-1755. [PMID: 29784586 DOI: 10.1016/j.ymthe.2018.04.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Revised: 04/18/2018] [Accepted: 04/20/2018] [Indexed: 02/01/2023] Open
Abstract
Ligament injuries occur frequently, substantially hindering routine daily activities and sports participation in patients. Surgical reconstruction using autogenous or allogeneic tissues is the gold standard treatment for ligament injuries. Although surgeons routinely perform ligament reconstructions, the integrity of these reconstructions largely depends on adequate biological healing of the interface between the ligament graft and the bone. We hypothesized that localized ultrasound-mediated, microbubble-enhanced therapeutic gene delivery to endogenous stem cells would lead to significantly improved ligament graft integration. To test this hypothesis, an anterior cruciate ligament reconstruction procedure was performed in Yucatan mini-pigs. A collagen scaffold was implanted in the reconstruction sites to facilitate recruitment of endogenous mesenchymal stem cells. Ultrasound-mediated reporter gene delivery successfully transfected 40% of cells recruited to the reconstruction sites. When BMP-6 encoding DNA was delivered, BMP-6 expression in the reconstruction sites was significantly enhanced. Micro-computed tomography and biomechanical analyses showed that ultrasound-mediated BMP-6 gene delivery led to significantly enhanced osteointegration in all animals 8 weeks after surgery. Collectively, these findings demonstrate that ultrasound-mediated gene delivery to endogenous mesenchymal progenitor cells can effectively improve ligament reconstruction in large animals, thereby addressing a major unmet orthopedic need and offering new possibilities for translation to the clinical setting.
Collapse
Affiliation(s)
- Maxim Bez
- Skeletal Biotech Laboratory, The Hebrew University-Hadassah Faculty of Dental Medicine, Ein Kerem, Jerusalem 91120, Israel; Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Thomas J Kremen
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Wafa Tawackoli
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Pablo Avalos
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Dmitriy Sheyn
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Galina Shapiro
- Skeletal Biotech Laboratory, The Hebrew University-Hadassah Faculty of Dental Medicine, Ein Kerem, Jerusalem 91120, Israel
| | - Joseph C Giaconi
- Department of Imaging, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Shiran Ben David
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jess G Snedeker
- Department of Orthopedics, University of Zurich, Zurich 8008, Switzerland
| | - Zulma Gazit
- Skeletal Biotech Laboratory, The Hebrew University-Hadassah Faculty of Dental Medicine, Ein Kerem, Jerusalem 91120, Israel; Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Katherine W Ferrara
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616, USA
| | - Dan Gazit
- Skeletal Biotech Laboratory, The Hebrew University-Hadassah Faculty of Dental Medicine, Ein Kerem, Jerusalem 91120, Israel; Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Gadi Pelled
- Skeletal Biotech Laboratory, The Hebrew University-Hadassah Faculty of Dental Medicine, Ein Kerem, Jerusalem 91120, Israel; Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| |
Collapse
|
21
|
Bez M, Sheyn D, Tawackoli W, Avalos P, Shapiro G, Giaconi JC, Da X, David SB, Gavrity J, Awad HA, Bae HW, Ley EJ, Kremen TJ, Gazit Z, Ferrara KW, Pelled G, Gazit D. In situ bone tissue engineering via ultrasound-mediated gene delivery to endogenous progenitor cells in mini-pigs. Sci Transl Med 2018; 9:9/390/eaal3128. [PMID: 28515335 DOI: 10.1126/scitranslmed.aal3128] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 02/08/2017] [Accepted: 04/14/2017] [Indexed: 12/20/2022]
Abstract
More than 2 million bone-grafting procedures are performed each year using autografts or allografts. However, both options carry disadvantages, and there remains a clear medical need for the development of new therapies for massive bone loss and fracture nonunions. We hypothesized that localized ultrasound-mediated, microbubble-enhanced therapeutic gene delivery to endogenous stem cells would induce efficient bone regeneration and fracture repair. To test this hypothesis, we surgically created a critical-sized bone fracture in the tibiae of Yucatán mini-pigs, a clinically relevant large animal model. A collagen scaffold was implanted in the fracture to facilitate recruitment of endogenous mesenchymal stem/progenitor cells (MSCs) into the fracture site. Two weeks later, transcutaneous ultrasound-mediated reporter gene delivery successfully transfected 40% of cells at the fracture site, and flow cytometry showed that 80% of the transfected cells expressed MSC markers. Human bone morphogenetic protein-6 (BMP-6) plasmid DNA was delivered using ultrasound in the same animal model, leading to transient expression and secretion of BMP-6 localized to the fracture area. Micro-computed tomography and biomechanical analyses showed that ultrasound-mediated BMP-6 gene delivery led to complete radiographic and functional fracture healing in all animals 6 weeks after treatment, whereas nonunion was evident in control animals. Collectively, these findings demonstrate that ultrasound-mediated gene delivery to endogenous mesenchymal progenitor cells can effectively treat nonhealing bone fractures in large animals, thereby addressing a major orthopedic unmet need and offering new possibilities for clinical translation.
Collapse
Affiliation(s)
- Maxim Bez
- Skeletal Biotech Laboratory, Hadassah Faculty of Dental Medicine, The Hebrew University of Jerusalem, Ein Kerem, Jerusalem 91120, Israel.,Department of Surgery, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Dmitriy Sheyn
- Department of Surgery, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA.,Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Wafa Tawackoli
- Department of Surgery, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA.,Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.,Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Pablo Avalos
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Galina Shapiro
- Skeletal Biotech Laboratory, Hadassah Faculty of Dental Medicine, The Hebrew University of Jerusalem, Ein Kerem, Jerusalem 91120, Israel
| | - Joseph C Giaconi
- Department of Imaging, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Xiaoyu Da
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Shiran Ben David
- Department of Surgery, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA.,Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jayne Gavrity
- Department of Biomedical Engineering and the Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Hani A Awad
- Department of Biomedical Engineering and the Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Hyun W Bae
- Department of Surgery, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Eric J Ley
- Department of Surgery, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Thomas J Kremen
- Department of Surgery, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA.,Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Zulma Gazit
- Skeletal Biotech Laboratory, Hadassah Faculty of Dental Medicine, The Hebrew University of Jerusalem, Ein Kerem, Jerusalem 91120, Israel.,Department of Surgery, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA.,Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.,Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Katherine W Ferrara
- Department of Biomedical Engineering, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616, USA
| | - Gadi Pelled
- Skeletal Biotech Laboratory, Hadassah Faculty of Dental Medicine, The Hebrew University of Jerusalem, Ein Kerem, Jerusalem 91120, Israel.,Department of Surgery, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA.,Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.,Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Dan Gazit
- Skeletal Biotech Laboratory, Hadassah Faculty of Dental Medicine, The Hebrew University of Jerusalem, Ein Kerem, Jerusalem 91120, Israel. .,Department of Surgery, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA.,Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.,Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.,Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
22
|
Delivery of the gene encoding the tumor suppressor Sef into prostate tumors by therapeutic-ultrasound inhibits both tumor angiogenesis and growth. Sci Rep 2017; 7:15060. [PMID: 29118380 PMCID: PMC5678190 DOI: 10.1038/s41598-017-12408-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 08/30/2017] [Indexed: 11/16/2022] Open
Abstract
Carcinomas constitute over 80% of all human cancer types with no effective therapy for metastatic disease. Here, we demonstrate, for the first time, the efficacy of therapeutic-ultrasound (TUS) to deliver a human tumor suppressor gene, hSef-b, to prostate tumors in vivo. Sef is downregulated in various human carcinomas, in a manner correlating with tumor aggressiveness. In vitro, hSef-b inhibited proliferation of TRAMP C2 cells and attenuated activation of ERK/MAPK and the master transcription factor NF-κB in response to FGF and IL-1/TNF, respectively. In vivo, transfection efficiency of a plasmid co-expressing hSef-b/eGFP into TRAMP C2 tumors was 14.7 ± 2.5% following a single TUS application. Repeated TUS treatments with hSef-b plasmid, significantly suppressed prostate tumor growth (60%) through inhibition of cell proliferation (60%), and reduction in blood vessel density (56%). In accordance, repeated TUS-treatments with hSef-b significantly inhibited in vivo expression of FGF2 and MMP-9. FGF2 is a known mitogen, and both FGF2/MMP-9 are proangiogenic factors. Taken together our results strongly suggest that hSef-b acts in a cell autonomous as well as non-cell autonomous manner. Moreover, the study demonstrates the efficacy of non-viral TUS-based hSef-b gene delivery approach for the treatment of prostate cancer tumors, and possibly other carcinomas where Sef is downregulated.
Collapse
|
23
|
Song Z, Wang Z, Shen J, Xu S, Hu Z. Nerve growth factor delivery by ultrasound-mediated nanobubble destruction as a treatment for acute spinal cord injury in rats. Int J Nanomedicine 2017; 12:1717-1729. [PMID: 28280337 PMCID: PMC5340249 DOI: 10.2147/ijn.s128848] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Spinal cord injuries (SCIs) can cause severe disability or death. Treatment options include surgical intervention, drug therapy, and stem cell transplantation. However, the efficacy of these methods for functional recovery remains unsatisfactory. Purpose This study was conducted to explore the effect of ultrasound (US)-mediated destruction of poly(lactic-co-glycolic acid) (PLGA) nanobubbles (NBs) expressing nerve growth factor (NGF) (NGF/PLGA NBs) on nerve regeneration in rats following SCI. Materials and methods Adult male Sprague Dawley rats were randomly divided into four treatment groups after Allen hit models of SCI were established. The groups were normal saline (NS) group, NGF and NBs group, NGF and US group, and NGF/PLGA NBs and US group. Histological changes after SCI were observed by hematoxylin and eosin staining. Neuron viability was determined by Nissl staining. Terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling staining was used to examine cell apoptosis. NGF gene and protein expressions were detected by quantitative reverse transcription polymerase chain reaction and Western blotting. Green fluorescent protein expression in the spinal cord was examined using an inverted fluorescence microscope. The recovery of neural function was determined using the Basso, Beattie, and Bresnahan test. Results NGF therapy using US-mediated NGF/PLGA NBs destruction significantly increased NGF expression, attenuated histological injury, decreased neuron loss, inhibited neuronal apoptosis in injured spinal cords, and increased BBB scores in rats with SCI. Conclusion US-mediated NGF/PLGA NBs destruction effectively transfects the NGF gene into target tissues and has a significant effect on the injured spinal cord. The combination of US irradiation and gene therapy through NGF/PLGA NBs holds great promise for the future of nanomedicine and the development of noninvasive treatment options for SCI and other diseases.
Collapse
Affiliation(s)
- Zhaojun Song
- Department of Orthopedics, The First Affiliated Hospital
| | - Zhigang Wang
- Institution of Ultrasound Imaging, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, People's Republic of China
| | - Jieliang Shen
- Department of Orthopedics, The First Affiliated Hospital
| | - Shengxi Xu
- Department of Orthopedics, The First Affiliated Hospital
| | - Zhenming Hu
- Department of Orthopedics, The First Affiliated Hospital
| |
Collapse
|
24
|
Ultrasound-Mediated Mesenchymal Stem Cells Transfection as a Targeted Cancer Therapy Platform. Sci Rep 2017; 7:42046. [PMID: 28169315 PMCID: PMC5294424 DOI: 10.1038/srep42046] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 01/05/2017] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) hold tremendous potential as a targeted cell-based delivery platform for inflammatory and cancer therapy. Genetic manipulation of MSCs, however, is challenging, and therefore, most studies using MSCs as therapeutic cell carriers have utilized viral vectors to transduce the cells. Here, we demonstrate, for the first time, an alternative approach for the efficient transfection of MSCs; therapeutic ultrasound (TUS). Using TUS with low intensities and moderate frequencies, MSCs were transfected with a pDNA encoding for PEX, a protein that inhibits tumor angiogenesis, and studied as a cell vehicle for in vivo tumor therapy. TUS application did not alter the MSCs' stemness or their homing capabilities, and the transfected MSCs transcribed biologically active PEX. Additionally, in a mouse model, 70% inhibition of prostate tumor growth was achieved following a single I.V. administration of MSCs that were TUS-transfected with pPEX. Further, the repeated I.V. administration of TUS-pPEX transfected-MSCs enhanced tumor inhibition up to 84%. Altogether, these results provide a proof of concept that TUS-transfected MSCs can be effectively used as a cell-based delivery approach for the prospective treatment of cancer.
Collapse
|
25
|
Xue Y, Yang G, Wang C, Li X, Du G. Effects of shRNA-Mediated SOX9 Inhibition on Cell Proliferation and Apoptosis in Human HCC Cell Line Hep3B Mediated by Ultrasound-Targeted Microbubble Destruction (UTMD). Cell Biochem Biophys 2017; 73:553-558. [PMID: 27352352 DOI: 10.1007/s12013-015-0685-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the most aggressive tumors in humans. The survival rate of patients is still very poor as current therapies offer limited treatment efficacy. Therefore, it is necessary to explore novel and more effective strategies to treat HCC. Recently, Ultrasound-targeted microbubble destruction (UTMD) has been shown to be a better alternative to viral vectors in delivering plasmid DNA into cells. In this study, we thus first determined the effect of combining UTMD with effectene on the transfection efficiency in human Hep3B cells. Transfection rate of the [effectene + shRNA-SOX9 + UTMD] group was the highest among the five groups, and were significantly higher than that of the [effectene + shRNA-SOX9] or [shRNA-SOX9 + UTMD] groups, while there was no significant difference between [shRNA-SOX9 alone] and [shRNA-SOX9 + UTMD] groups. Expression of SOX9 mRNA and protein was the lowest in effectene + shRNA-SOX9 + UTMD group. Moreover, transfection of shRNA-SOX9 with UTMD and effectene in combination could markedly inhibit the proliferation and induced cell apoptosis of Hep3B cells. These results suggest that the efficiency of gene delivery is remarkably increased when UTMD is combined with other transfection strategies, such as effectene. In conclusion, our research demonstrates that combining conventional transfection methods with UTMD achieves better transfection efficiency and that this can provide an improved gene delivery system for gene therapy.
Collapse
Affiliation(s)
- Yan Xue
- Department of Ultrasound, Linyi People's Hospital Affiliated to Shandong University, Linyi, 276003, People's Republic of China
| | - Guilun Yang
- Department of Medical Imaging, Linyi People's Hospital Affiliated to Shandong University, Linyi, 276003, People's Republic of China.
| | - Changmei Wang
- Department of Healthcare, Shandong University Affiliated Jinan Central Hospital, Jinan, 250013, People's Republic of China
| | - Xiaodong Li
- Department of Medical Imaging, Linyi People's Hospital Affiliated to Shandong University, Linyi, 276003, People's Republic of China
| | - Guijin Du
- Department of Medical Imaging, Linyi People's Hospital Affiliated to Shandong University, Linyi, 276003, People's Republic of China
| |
Collapse
|
26
|
Chiper M, Tounsi N, Kole R, Kichler A, Zuber G. Self-aggregating 1.8kDa polyethylenimines with dissolution switch at endosomal acidic pH are delivery carriers for plasmid DNA, mRNA, siRNA and exon-skipping oligonucleotides. J Control Release 2016; 246:60-70. [PMID: 27956144 DOI: 10.1016/j.jconrel.2016.12.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 11/23/2016] [Accepted: 12/05/2016] [Indexed: 11/24/2022]
Abstract
Efficiency of polyethylenimine (PEI) for nucleic acid delivery is affected by the size of the carrier and length of the nucleic acids. For instance, PEIs with molecular weights between 10-30kDa provide optimal DNA delivery activity whereas PEIs with molecular weights below 1.8kDa are ineffective. The activity of PEI is also severely diminished by substitution of DNA for shorter nucleic acids such as mRNA or siRNA. Here, through chemical modification of the primary amines to aromatic domains we achieved nucleic acid delivery by the 1.8kDa polyethylenimine (PEI) particles. This modification did not affect the PEI buffering abilities but enhanced its pH-sensitive aggregation, enabling stabilization of the polyplex outside the cell while still allowing nucleic acid release following cellular entry. The aromatic PEIs were then evaluated for their gene, mRNA, siRNA and 2'O-methyl phosphorothioate oligonucleotide in vitro transfection abilities. The salicylamide-grafted PEI showed to be a reliable carrier for delivering nucleic acids with cytoplasmic activity such as the mRNA and siRNA or nuclear diffusible oligonucleotide. It was then further equipped with polyethyleneglycol (PEG) and the delivery efficiency of the copolymer was tested in vivo for regeneration of dystrophin in the muscle of mdx mouse through a 2'O-methyl phosphorothioate-mediated splicing modulation. Intramuscular administration of polyplexes resulted in dystrophin-positive fibers in a mouse model of Duchenne muscular dystrophy without apparent toxicity. These findings indicate that precise modifications of low molecular weight PEI improve its bio-responsiveness and yield delivery vehicles for nucleic acids of various types in vitro and in vivo.
Collapse
Affiliation(s)
- Manuela Chiper
- Molecular and Pharmaceutical Engineering of Biologics, CNRS - Université de Strasbourg UMR 7242, Boulevard Sebastien Brant, 67412 Illkirch, France; Faculté de Pharmacie - Université de Strasbourg, 74 Route du Rhin, F-67400 Illkirch, France
| | - Nassera Tounsi
- Faculté de Pharmacie - Université de Strasbourg, 74 Route du Rhin, F-67400 Illkirch, France; Laboratory of Therapeutic Innovation UMR 7200, CNRS - Université de Strasbourg, France
| | - Ryszard Kole
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC 27599, United States
| | - Antoine Kichler
- Faculté de Pharmacie - Université de Strasbourg, 74 Route du Rhin, F-67400 Illkirch, France; Laboratoire de Conception et d'Application de Molécules Bioactives, CNRS - Université de Strasbourg UMR 7199, France; Genethon, 91000 Evry, France
| | - Guy Zuber
- Molecular and Pharmaceutical Engineering of Biologics, CNRS - Université de Strasbourg UMR 7242, Boulevard Sebastien Brant, 67412 Illkirch, France.
| |
Collapse
|
27
|
Tamošiūnas M, Kadikis R, Saknite I, Baltušnikas J, Kilikevičius A, Lihachev A, Petrovska R, Jakovels D, Šatkauskas S. Noninvasive optical diagnostics of enhanced green fluorescent protein expression in skeletal muscle for comparison of electroporation and sonoporation efficiencies. JOURNAL OF BIOMEDICAL OPTICS 2016; 21:45003. [PMID: 27129126 DOI: 10.1117/1.jbo.21.4.045003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 04/12/2016] [Indexed: 06/05/2023]
Abstract
We highlight the options available for noninvasive optical diagnostics of reporter gene expression in mouse tibialis cranialis muscle. An in vivo multispectral imaging technique combined with fluorescence spectroscopy point measurements has been used for the transcutaneous detection of enhanced green fluorescent protein (EGFP) expression, providing information on location and duration of EGFP expression and allowing quantification of EGFP expression levels. For EGFP coding plasmid (pEGFP-Nuc Vector, 10 μg/50 ml 10 μg/50 ml ) transfection, we used electroporation or ultrasound enhanced microbubble cavitation [sonoporation (SP)]. The transcutaneous EGFP fluorescence in live mice was monitored over a period of one year using the described parameters: area of EGFP positive fibers, integral intensity, and mean intensity of EGFP fluorescence. The most efficient transfection of EGFP coding plasmid was achieved, when one high voltage and four low voltage electric pulses were applied. This protocol resulted in the highest short-term and long-term EGFP expression. Other electric pulse protocols as well as SP resulted in lower fluorescence intensities of EGFP in the transfected area. We conclude that noninvasive multispectral imaging technique combined with fluorescence spectroscopy point measurements is a suitable method to estimate the dynamics and efficiency of reporter gene transfection in vivo.
Collapse
Affiliation(s)
- Mindaugas Tamošiūnas
- Vytautas Magnus University, Biophysical Research Group, Faculty of Natural Sciences, Vileikos 8, Kaunas LT-44404, LithuaniabVytautas Magnus University, Department of Biochemistry, Faculty of Natural Sciences, Vileikos 8, Kaunas LT-44404, Lithuania
| | - Roberts Kadikis
- Institute of Electronics and Computer Science, 14 Dzerbenes Street, Riga LV-1006, Latvia
| | - Inga Saknite
- University of Latvia, Institute of Atomic Physics and Spectroscopy, 19 Rainis Boulevard, Riga LV-1586, Latvia
| | - Juozas Baltušnikas
- Lithuanian Sports University, Institute of Sports Sciences and Innovation, Sporto 6, LT-44221 Kaunas, Lithuania
| | - Audrius Kilikevičius
- Lithuanian Sports University, Institute of Sports Sciences and Innovation, Sporto 6, LT-44221 Kaunas, Lithuania
| | - Alexey Lihachev
- University of Latvia, Institute of Atomic Physics and Spectroscopy, 19 Rainis Boulevard, Riga LV-1586, Latvia
| | - Ramona Petrovska
- Latvian Biomedical Research and Study Centre, Ratsupites iela 1, Riga LV-1067, Latvia
| | - Dainis Jakovels
- University of Latvia, Institute of Atomic Physics and Spectroscopy, 19 Rainis Boulevard, Riga LV-1586, Latvia
| | - Saulius Šatkauskas
- Vytautas Magnus University, Biophysical Research Group, Faculty of Natural Sciences, Vileikos 8, Kaunas LT-44404, Lithuania
| |
Collapse
|
28
|
Wang H, Gauthier M, Kelly JR, Miller RJ, Xu M, O'Brien WD, Cheng J. Targeted Ultrasound-Assisted Cancer-Selective Chemical Labeling and Subsequent Cancer Imaging using Click Chemistry. Angew Chem Int Ed Engl 2016; 55:5452-6. [PMID: 27010510 PMCID: PMC4918225 DOI: 10.1002/anie.201509601] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 01/29/2016] [Indexed: 01/01/2023]
Abstract
Metabolic sugar labeling followed by the use of reagent-free click chemistry is an established technique for in vitro cell targeting. However, selective metabolic labeling of the target tissues in vivo remains a challenge to overcome, which has prohibited the use of this technique for targeted in vivo applications. Herein, we report the use of targeted ultrasound pulses to induce the release of tetraacetyl N-azidoacetylmannosamine (Ac4 ManAz) from microbubbles (MBs) and its metabolic expression in the cancer area. Ac4 ManAz-loaded MBs showed great stability under physiological conditions, but rapidly collapsed in the presence of tumor-localized ultrasound pulses. The released Ac4 ManAz from MBs was able to label 4T1 tumor cells with azido groups and significantly improved the tumor accumulation of dibenzocyclooctyne (DBCO)-Cy5 by subsequent click chemistry. We demonstrated for the first time that Ac4 ManAz-loaded MBs coupled with the use of targeted ultrasound could be a simple but powerful tool for in vivo cancer-selective labeling and targeted cancer therapies.
Collapse
Affiliation(s)
- Hua Wang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, USA
| | - Marianne Gauthier
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Jamie R Kelly
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Rita J Miller
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Ming Xu
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, USA
| | - William D O'Brien
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
| | - Jianjun Cheng
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, USA.
| |
Collapse
|
29
|
Wang H, Gauthier M, Kelly JR, Miller RJ, Xu M, O'Brien WD, Cheng J. Targeted Ultrasound‐Assisted Cancer‐Selective Chemical Labeling and Subsequent Cancer Imaging using Click Chemistry. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201509601] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Hua Wang
- Department of Materials Science and Engineering University of Illinois at Urbana-Champaign USA
| | - Marianne Gauthier
- Department of Electrical and Computer Engineering University of Illinois at Urbana-Champaign Urbana IL 61801 USA
| | - Jamie R. Kelly
- Department of Electrical and Computer Engineering University of Illinois at Urbana-Champaign Urbana IL 61801 USA
| | - Rita J. Miller
- Department of Electrical and Computer Engineering University of Illinois at Urbana-Champaign Urbana IL 61801 USA
| | - Ming Xu
- Department of Materials Science and Engineering University of Illinois at Urbana-Champaign USA
| | - William D. O'Brien
- Department of Electrical and Computer Engineering University of Illinois at Urbana-Champaign Urbana IL 61801 USA
| | - Jianjun Cheng
- Department of Materials Science and Engineering University of Illinois at Urbana-Champaign USA
| |
Collapse
|
30
|
Wen WS, Yuan ZM, Ma SJ, Xu J, Yuan DT. CRISPR-Cas9 systems: versatile cancer modelling platforms and promising therapeutic strategies. Int J Cancer 2016; 138:1328-36. [PMID: 26044706 DOI: 10.1002/ijc.29626] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 05/12/2015] [Accepted: 05/28/2015] [Indexed: 12/26/2022]
Abstract
The RNA-guided nuclease CRISPR-Cas9 (clustered regularly interspaced short palindromic repeats-CRISPR associated nuclease 9) and its variants such as nickase Cas9, dead Cas9, guide RNA scaffolds and RNA-targeting Cas9 are convenient and versatile platforms for site-specific genome editing and epigenome modulation. They are easy-to-use, simple-to-design and capable of targeting multiple loci simultaneously. Given that cancer develops from cumulative genetic and epigenetic alterations, CRISPR-Cas9 and its variants (hereafter referred to as CRISPR-Cas9 systems) hold extensive application potentials in cancer modeling and therapy. To date, they have already been applied to model oncogenic mutations in cell lines (e.g., Choi and Meyerson, Nat Commun 2014;5:3728) and in adult animals (e.g., Xue et al., Nature 2014;514:380-4), as well as to combat cancer by disabling oncogenic viruses (e.g., Hu et al., Biomed Res Int 2014;2014:612823) or by manipulating cancer genome (e.g., Liu et al., Nat Commun 2014;5:5393). Given the importance of epigenome and transcriptome in tumourigenesis, manipulation of cancer epigenome and transcriptome for cancer modeling and therapy is a promising area in the future. Whereas (epi)genetic modifications of cancer microenvironment with CRISPR-Cas9 systems for therapeutic purposes represent another promising area in cancer research. Herein, we introduce the functions and mechanisms of CRISPR-Cas9 systems in genome editing and epigenome modulation, retrospect their applications in cancer modelling and therapy, discuss limitations and possible solutions and propose future directions, in hope of providing concise and enlightening information for readers interested in this area.
Collapse
Affiliation(s)
- Wan-Shun Wen
- Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang Province, China
| | - Zhi-Min Yuan
- Cervical Disease Clinic, Jiangsu Huai'an Maternity and Children Hospital, Huai'an, China
| | - Shi-Jie Ma
- Department of Gastroenterology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, China
| | - Jiang Xu
- Department of Rehabilitation, the Affiliated Huai'an Hospital of Xuzhou Medical College and the Second People's Hospital of Huai'an, Huai'an, China
| | - Dong-Tang Yuan
- Department of Orthopedics, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China
| |
Collapse
|
31
|
Shapiro G, Wong AW, Bez M, Yang F, Tam S, Even L, Sheyn D, Ben-David S, Tawackoli W, Pelled G, Ferrara KW, Gazit D. Multiparameter evaluation of in vivo gene delivery using ultrasound-guided, microbubble-enhanced sonoporation. J Control Release 2015; 223:157-164. [PMID: 26682505 DOI: 10.1016/j.jconrel.2015.12.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Revised: 10/27/2015] [Accepted: 12/01/2015] [Indexed: 10/22/2022]
Abstract
More than 1800 gene therapy clinical trials worldwide have targeted a wide range of conditions including cancer, cardiovascular diseases, and monogenic diseases. Biological (i.e. viral), chemical, and physical approaches have been developed to deliver nucleic acids into cells. Although viral vectors offer the greatest efficiency, they also raise major safety concerns including carcinogenesis and immunogenicity. The goal of microbubble-mediated sonoporation is to enhance the uptake of drugs and nucleic acids. Insonation of microbubbles is thought to facilitate two mechanisms for enhanced uptake: first, deflection of the cell membrane inducing endocytotic uptake, and second, microbubble jetting inducing the formation of pores in the cell membrane. We hypothesized that ultrasound could be used to guide local microbubble-enhanced sonoporation of plasmid DNA. With the aim of optimizing delivery efficiency, we used nonlinear ultrasound and bioluminescence imaging to optimize the acoustic pressure, microbubble concentration, treatment duration, DNA dosage, and number of treatments required for in vivo Luciferase gene expression in a mouse thigh muscle model. We found that mice injected with 50μg luciferase plasmid DNA and 5×10(5) microbubbles followed by ultrasound treatment at 1.4MHz, 200kPa, 100-cycle pulse length, and 540 Hz pulse repetition frequency (PRF) for 2min exhibited superior transgene expression compared to all other treatment groups. The bioluminescent signal measured for these mice on Day 4 post-treatment was 100-fold higher (p<0.0001, n=5 or 6) than the signals for controls treated with DNA injection alone, DNA and microbubble injection, or DNA injection and ultrasound treatment. Our results indicate that these conditions result in efficient gene delivery and prolonged gene expression (up to 21days) with no evidence of tissue damage or off-target delivery. We believe that these promising results bear great promise for the development of microbubble-enhanced sonoporation-induced gene therapies.
Collapse
Affiliation(s)
- Galina Shapiro
- Skeletal Biotech Laboratory, The Hebrew University-Hadassah Faculty of Dental Medicine, Ein Kerem, Jerusalem 91120, Israel
| | - Andrew W Wong
- University of California, Davis, Department of Biomedical Engineering, 451 Health Sciences Drive, Davis, CA 95616, USA
| | - Maxim Bez
- Skeletal Biotech Laboratory, The Hebrew University-Hadassah Faculty of Dental Medicine, Ein Kerem, Jerusalem 91120, Israel
| | - Fang Yang
- University of California, Davis, Department of Biomedical Engineering, 451 Health Sciences Drive, Davis, CA 95616, USA
| | - Sarah Tam
- University of California, Davis, Department of Biomedical Engineering, 451 Health Sciences Drive, Davis, CA 95616, USA
| | - Lisa Even
- University of California, Davis, Department of Biomedical Engineering, 451 Health Sciences Drive, Davis, CA 95616, USA
| | - Dmitriy Sheyn
- Department of Surgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA; Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Shiran Ben-David
- Department of Surgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA; Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Wafa Tawackoli
- Department of Surgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA; Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Gadi Pelled
- Skeletal Biotech Laboratory, The Hebrew University-Hadassah Faculty of Dental Medicine, Ein Kerem, Jerusalem 91120, Israel; Department of Surgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA; Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Katherine W Ferrara
- University of California, Davis, Department of Biomedical Engineering, 451 Health Sciences Drive, Davis, CA 95616, USA
| | - Dan Gazit
- Skeletal Biotech Laboratory, The Hebrew University-Hadassah Faculty of Dental Medicine, Ein Kerem, Jerusalem 91120, Israel; Department of Surgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA; Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| |
Collapse
|
32
|
Tavri S, Vezeridis A, Cui W, Mattrey RF. In Vivo Transfection and Detection of Gene Expression of Stem Cells Preloaded with DNA-carrying Microbubbles. Radiology 2015; 276:518-25. [PMID: 25811427 DOI: 10.1148/radiol.15141380] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
PURPOSE To determine whether (a) stem cells loaded with DNA-carrying microbubbles (MBs) can be transfected in vivo, (b) the cells remain alive to express the gene, and (c) gene expression is sufficiently robust to be detected in vivo. MATERIALS AND METHODS The study was approved by the Institutional Animal Care and Use Committee. Cationic MBs were prepared, characterized, and loaded with pLuciferase green fluorescent protein (GFP) plasmid. Loading was confirmed with SYBR Gold staining (Life Technologies, Carlsbad, Calif). C17.2 cells were loaded with the DNA-carrying MBs. Two hundred thousand cells suspended in 20 μL phosphate-buffered saline were mixed with 200 μL Matrigel (BD Biosciences, San Jose, Calif) and injected in both flanks of eight nude mice. One of the Matrigel (BD Biosciences) injections contained 50 000 cells pretransfected in vitro by using lipofectamine as a positive control. Nine flanks were exposed to 2.25-MHz ultrasonic pulses at 50% duty cycle for 1 minute at 1 W/cm(2) (n = 3) or 2 W/cm(2) (n = 6), and six flanks served as the negative control. Two days later, bioluminescent images were acquired in each mouse every 3 minutes for 1 hour after the intraperitoneal injection of d-luciferin (Perkin Elmer, Waltham, Mass). Differences between groups were assessed by using the nonparametric Kruskal-Wallis test with Wilcoxon rank sum tests for follow-up comparisons. Mice were then killed, plugs were explanted, and alternate sections were stained with hematoxylin-eosin or stained for GFP expression. RESULTS Mean DNA-loaded MB diameter ± standard deviation was 2.87 μm ± 1.69 with the DNA associated with the MB shell. C17.2 cells were associated with 2-4 MBs each, and more than 90% were viable. Peak background subtracted bioluminescent signal was fourfold higher when cells were exposed to 2 W/cm(2) pulses as compared with 1 W/cm(2) pulses (P = .02) and negative controls (P = .002). Histologic examination showed cells within the Matrigel (BD Biosciences) with robust GFP expression only after 2 W/cm(2) ultrasound exposure and lipofectamine transfection. CONCLUSION Stem cells loaded with DNA-carrying MBs can be transfected in vivo with ultrasonic pulses and remain alive to demonstrate robust gene expression.
Collapse
Affiliation(s)
- Sidhartha Tavri
- From the Department of Radiology, University of California-San Diego, 200 W Arbor Dr, San Diego, CA 92103
| | - Alexander Vezeridis
- From the Department of Radiology, University of California-San Diego, 200 W Arbor Dr, San Diego, CA 92103
| | - Wenjin Cui
- From the Department of Radiology, University of California-San Diego, 200 W Arbor Dr, San Diego, CA 92103
| | - Robert F Mattrey
- From the Department of Radiology, University of California-San Diego, 200 W Arbor Dr, San Diego, CA 92103
| |
Collapse
|
33
|
Abstract
The key impediment to the successful application of gene therapy in clinics is not the paucity of therapeutic genes. It is rather the lack of nontoxic and efficient strategies to transfer therapeutic genes into target cells. Over the past few decades, considerable progress has been made in gene transfer technologies, and thus far, three different delivery systems have been developed with merits and demerits characterizing each system. Viral and chemical methods of gene transfer utilize specialized carrier to overcome membrane barrier and facilitate gene transfer into cells. Physical methods, on the other hand, utilize various forms of mechanical forces to enforce gene entry into cells. Starting in 1980s, physical methods have been introduced as alternatives to viral and chemical methods to overcome various extra- and intracellular barriers that limit the amount of DNA reaching the intended cells. Accumulating evidence suggests that it is quite feasible to directly translocate genes into cytoplasm or even nuclei of target cells by means of mechanical force, bypassing endocytosis, a common pathway for viral and nonviral vectors. Indeed, several methods have been developed, and the majority of them share the same underlying mechanism of gene transfer, i.e., physically created transient pores in cell membrane through which genes get into cells. Here, we provide an overview of the current status and future research directions in the field of physical methods of gene transfer.
Collapse
|
34
|
Kilroy JP, Klibanov AL, Wamhoff BR, Bowles DK, Hossack JA. Localized in vivo model drug delivery with intravascular ultrasound and microbubbles. ULTRASOUND IN MEDICINE & BIOLOGY 2014; 40:2458-67. [PMID: 25130449 PMCID: PMC4400670 DOI: 10.1016/j.ultrasmedbio.2014.04.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 04/08/2014] [Accepted: 04/10/2014] [Indexed: 05/19/2023]
Abstract
An intravascular ultrasound (IVUS) and microbubble drug delivery system was evaluated in both ex vivo and in vivo swine vessel models. Microbubbles with the fluorophore DiI embedded in the shell as a model drug were infused into ex vivo swine arteries at a physiologic flow rate (105 mL/min) while a 5-MHz IVUS transducer applied ultrasound. Ultrasound pulse sequences consisted of acoustic radiation force pulses to displace DiI-loaded microbubbles from the vessel lumen to the wall, followed by higher-intensity delivery pulses to release DiI into the vessel wall. Insonation with both the acoustic radiation force pulse and the delivery pulse increased DiI deposition 10-fold compared with deposition with the delivery pulse alone. Localized delivery of DiI was then demonstrated in an in vivo swine model. The theoretical transducer beam width predicted the measured angular extent of delivery to within 11%. These results indicate that low-frequency IVUS catheters are a viable method for achieving localized drug delivery with microbubbles.
Collapse
Affiliation(s)
- Joseph P Kilroy
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Alexander L Klibanov
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA; Cardiovascular Division, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Brian R Wamhoff
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA; Cardiovascular Division, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA; Hemoshear, LLC, Charlottesville, Virginia, USA
| | - Douglas K Bowles
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, USA
| | - John A Hossack
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA.
| |
Collapse
|
35
|
Yoshida K, Obata K, Tsukamoto A, Ushida T, Watanabe Y. Limited damage of tissue mimic caused by a collapsing bubble under low-frequency ultrasound exposure. ULTRASONICS 2014; 54:1603-1609. [PMID: 24751130 DOI: 10.1016/j.ultras.2014.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 02/27/2014] [Accepted: 03/21/2014] [Indexed: 06/03/2023]
Abstract
In this study, we investigated the bubble induced serious damage to tissue mimic exposed to 27-kHz ultrasound. The initial bubble radius ranged from 80 to 100 μm, which corresponded approximately to the experimentally-evaluated resonant radius of the given ultrasound frequency. The tissue mimic consisted of 10 wt% gelatine gel covered with cultured canine kidney epithelial cells. The collapsing bubble behaviour during the ultrasound exposure with negative peak pressures of several hundred kPa was captured by a high-speed camera system. After ultrasound exposure, a cell viability test was conducted based on microscopic bright-field images and fluorescence images for living and dead cells. In the viability test, cells played a role in indicating the damaged area. The bubble oscillations killed the cells, and on occasion detached layers of cultured cells from the gel. The damaged area was comparable or slightly larger than the initial bubble size, and smaller than the maximum bubble size. We concluded that only a small area in close proximity to the bubble could be damaged even above transient cavitation threshold.
Collapse
Affiliation(s)
- Kenji Yoshida
- Laboratory of Ultrasonic Electronics, Doshisha University, 610-0321 Kyotanabe, Kyoto, Japan; Center for Frontier Medical Engineering, Chiba University, 263-8522 Chiba, Japan.
| | - Kazuya Obata
- Laboratory of Ultrasonic Electronics, Doshisha University, 610-0321 Kyotanabe, Kyoto, Japan
| | - Akira Tsukamoto
- Department of Applied Physics, National Defense Academy of Japan, 239-8686 Yokosuka, Kanagawa, Japan
| | - Takashi Ushida
- Center for Disease Biology and Integrative Medicine, The University of Tokyo, 113-0033 Bunkyo-ku, Tokyo, Japan
| | - Yoshiaki Watanabe
- Laboratory of Ultrasonic Electronics, Doshisha University, 610-0321 Kyotanabe, Kyoto, Japan
| |
Collapse
|
36
|
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: 91] [Impact Index Per Article: 9.1] [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
|
37
|
Lo CW, Chang Y, Lee JL, Tsai WB, Chen WS. Tertiary-amine functionalized polyplexes enhanced cellular uptake and prolonged gene expression. PLoS One 2014; 9:e97627. [PMID: 24827929 PMCID: PMC4020921 DOI: 10.1371/journal.pone.0097627] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 04/20/2014] [Indexed: 12/31/2022] Open
Abstract
Ultrasound (US) has been found to facilitate the transport of DNA across cell membranes. However, the transfection efficiency is generally low, and the expression duration of the transfected gene is brief. In this study, a tertiary polycation, Poly(2-(dimethylamino) ethyl methacrylate) (PDMAEMA), was used as a carrier for US-mediated gene transfection. Its in-vitro and in-vivo effects on the transfection efficiency and the expression duration were evaluated. A mixture of pCI-neo-luc and PDMAEMA was transfected to cultured cells or mouse muscle by exposure to 1-MHz pulse US. A strong expression of luciferase was found 10 days after the transfection in vitro regardless of US exposure. However, effective transfection only occurred in the US groups in vivo. The transfection ability depended on the weight ratio of PDMAEMA to DNA, and was different for the in-vitro and in-vivo conditions. Lower weight ratios, e.g., 0.25, exhibited better in-vivo expression for at least 45 days.
Collapse
Affiliation(s)
- Chia-Wen Lo
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan, ROC
| | - Yung Chang
- R&D Center for Membrane Technology and Department of Chemical Engineering, Chung Yuan Christian University, Jhong-Li, Taoyuan, Taiwan, ROC
| | - Jyun-Lin Lee
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan, ROC
| | - Wei-Bor Tsai
- Department of Chemical Engineering, National Taiwan University College of Medicine, Taipei, Taiwan, ROC
| | - Wen-Shiang Chen
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan, ROC
- * E-mail:
| |
Collapse
|
38
|
Polyxylitol-based gene carrier improves the efficiency of gene transfer through enhanced endosomal osmolysis. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2014; 10:525-34. [DOI: 10.1016/j.nano.2013.10.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2013] [Revised: 08/07/2013] [Accepted: 10/14/2013] [Indexed: 11/17/2022]
|
39
|
Lee JL, Lo CW, Inserra C, Béra JC, Chen WS. Ultrasound enhanced PEI-mediated gene delivery through increasing the intracellular calcium level and PKC-δ protein expression. Pharm Res 2014; 31:2354-66. [PMID: 24623478 DOI: 10.1007/s11095-014-1332-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 02/08/2014] [Indexed: 12/16/2022]
Abstract
PURPOSE Polyethylenimine (PEI), a cationic polymer, has been shown to aggregate plasmid DNA and facilitate its internalization. It has also been shown that combining ultrasound (US) with PEI could enhance and prolong in vitro and in vivo transgene expression. However, the role US in the enhancement of PEI uptake is poorly understood. This study investigates the impact of US on PEI-mediated gene transfection. METHODS Specific endocytosis pathway siRNA, including clathrin HC siRNA, caveolin-1 siRNA and protein kinase C-delta (PKC-δ) siRNA, are used to block the corresponding endocytosis pathways prior to the transfection of luciferase DNA/PEI polyplexes to cultured cells by 1-MHz pulsed US with ultrasound contrast agent SonoVue®. RESULTS Transgene expression was found not to be enhanced by US treatment in the presence of the PKC-δ siRNA. We further demonstrated that PKC-δ protein could be enhanced at 6 h after US exposure. Moreover, intracellular calcium levels were found to be significantly increased at 3 h after US exposure, while transgene expressions were significantly reduced in the presence of calcium channel blockers both in vitro and in vivo. CONCLUSIONS Our results suggest that US enhanced PEI-mediated gene transfection specifically by increasing PKC-δ related fluid phase endocytosis, which was induced by increasing the intracellular calcium levels.
Collapse
Affiliation(s)
- Jyun-Lin Lee
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital National Taiwan University College of Medicine, Taipei, Taiwan, Republic of China
| | | | | | | | | |
Collapse
|
40
|
Ultrasound-enhanced delivery of morpholino with Bubble liposomes ameliorates the myotonia of myotonic dystrophy model mice. Sci Rep 2014; 3:2242. [PMID: 23873129 PMCID: PMC3718203 DOI: 10.1038/srep02242] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Accepted: 06/21/2013] [Indexed: 11/08/2022] Open
Abstract
Phosphorodiamidate morpholino oligonucleotide (PMO)-mediated control of the alternative splicing of the chloride channel 1 (CLCN1) gene is a promising treatment for myotonic dystrophy type 1 (DM1) because the abnormal splicing of this gene causes myotonia in patients with DM1. In this study, we optimised a PMO sequence to correct Clcn1 alternative splicing and successfully remedied the myotonic phenotype of a DM1 mouse model, the HSALR mouse. To enhance the efficiency of delivery of PMO into HSALR mouse muscles, Bubble liposomes, which have been used as a gene delivery tool, were applied with ultrasound exposure. Effective delivery of PMO led to increased expression of Clcn1 protein in skeletal muscle and the amelioration of myotonia. Thus, PMO-mediated control of the alternative splicing of the Clcn1 gene must be important target of antisense therapy of DM1.
Collapse
|
41
|
Introduction of Genes via Sonoporation and Electroporation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 818:231-54. [DOI: 10.1007/978-1-4471-6458-6_12] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
42
|
Kósa M, Zádor E. Transfection efficiency along the regenerating soleus muscle of the rat. Mol Biotechnol 2013; 54:220-7. [PMID: 22580850 DOI: 10.1007/s12033-012-9555-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
We investigated the efficiency of a single plasmid transfection along the longitudinal axis of the regenerating soleus of young rats. This also reflected transfection efficiency along the fibers because the soleus is a nearly fusiform muscle in young animals. The complete regeneration was induced by notexin and the transfection was made by intramuscular injection of enhanced green fluorescent protein- or Discosoma red-coding plasmids after 4 days. One week after transfection the number of transfected fibers was higher at the place of injection (i.e., in the muscle belly) and lower or absent at the ends of the muscle. The inspection of longitudinal sections and neuromuscular endplates indicated that one of the reasons of uneven transfection might be the shortness of transfected myotubes and the other reason might be the limit of diffusion of transgenic proteins from the expressing nuclei. As a result, the efficiency of transfection in the whole regenerating muscle was much lower than it could be estimated from the most successfully transfected part.
Collapse
Affiliation(s)
- Magdolna Kósa
- Department of Biochemistry, University of Szeged, Szeged, Hungary.
| | | |
Collapse
|
43
|
Azab BM, Dash R, Das SK, Bhutia SK, Sarkar S, Shen XN, Quinn BA, Dent P, Dmitriev IP, Wang XY, Curiel DT, Pellecchia M, Reed JC, Sarkar D, Fisher PB. Enhanced prostate cancer gene transfer and therapy using a novel serotype chimera cancer terminator virus (Ad.5/3-CTV). J Cell Physiol 2013; 229:34-43. [PMID: 23868767 DOI: 10.1002/jcp.24408] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Accepted: 05/14/2013] [Indexed: 12/27/2022]
Abstract
Few options are available for treating patients with advanced prostate cancer (PC). As PC is a slow growing disease and accessible by ultrasound, gene therapy could provide a viable option for this neoplasm. Conditionally replication-competent adenoviruses (CRCAs) represent potentially useful reagents for treating PC. We previously constructed a CRCA, cancer terminator virus (CTV), which showed efficacy both in vitro and in vivo for PC. The CTV was generated on a serotype 5-background (Ad.5-CTV) with infectivity depending on Coxsackie-Adenovirus Receptors (CARs). CARs are frequently reduced in many tumor types, including PCs thereby limiting effective Ad-mediated therapy. Using serotype chimerism, a novel CTV (Ad.5/3-CTV) was created by replacing the Ad.5 fiber knob with the Ad.3 fiber knob thereby facilitating infection in a CAR-independent manner. We evaluated Ad.5/3-CTV in comparison with Ad.5-CTV in low CAR human PC cells, demonstrating higher efficiency in inhibiting cell viability in vitro. Moreover, Ad.5/3-CTV potently suppressed in vivo tumor growth in a nude mouse xenograft model and in a spontaneously induced PC that develops in Hi-myc transgenic mice. Considering the significant responses in a Phase I clinical trial of a non-replicating Ad.5-mda-7 in advanced cancers, Ad.5/3-CTV may exert improved therapeutic benefit in a clinical setting.
Collapse
Affiliation(s)
- Belal M Azab
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Elmadbouh I, Haider HK, Ashraf M, Chachques JC. Preconditioning of Human Skeletal Myoblast with Stromal Cell-derived Factor-1α Promotes Cytoprotective Effects against Oxidative and Anoxic Stress. Int J Stem Cells 2013; 4:50-60. [PMID: 24298334 DOI: 10.15283/ijsc.2011.4.1.50] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2011] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND AND OBJECTIVES The incidence of human autologous transplanted skeletal myoblast (SkM) cell death in ischemic myocardium was higher in the first few days after cell therapy. We proposed that human SkM treated by human stromal cell-derived factor (SDF-1α) protein or tranfected by SDF-1α, precondition them against oxidative or anoxic injury. METHODS AND RESULTS The purification of human SkM (80∼90%) culture was assessed for desmin and CXCR4 expression using immunostaining and flow cytometry respectively. Cells were transfected to overexpress SDF-1α or treated with rSDF-1α (10∼200 ng/ml, 1∼4 h) were either exposed to anoxia or treated with 100μM H2O2 for different time periods (1∼6 h anoxia) (1∼3 h H2O2). Optimized conditions for transfection of SDF-1α gene into human SkM were achieved, using FuGene(TM)6/phSDF-1α(3:2 v/w, 4 h transfection) with 125μ M ZnCl2 (p< 0.001), up to 7 days post-transfection as compared with transfected SkM without ZnCl2 and non-transfected control cells. Transfection efficiency was assessed by immunostaining, ELISA, western blots and PCR. LDH analysis showed significant decrease in release of LDH after exposure to 6 h anoxia or 100μ M H2O2 for 2 h as compared with the normal un-treated or un-transfected SkM (p< 0.001). In western blots assay, SDF-1α over-expressing human SkM or treated with rSDF-1α induced marked expression of total Akt (1.2-fold) and phospho-Akt (2.7-fold), Bcl2 (1.6-fold) and VEGF (5.8-fold) after exposure to 6 h anoxia as compared with human SkM controls. CONCLUSIONS The preconditioning of donor transplanted human SkM with SDF-1α increased cell survival and promoted cytoprotective effect against oxidative or anoxic injury that may be an innovative approach for clinical application.
Collapse
Affiliation(s)
- Ibrahim Elmadbouh
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Ohio, USA ; Department of Cardiovascular Surgery (LBR), Pompidou Hospital, Paris-5 University, Paris, France ; Faculty of Medicine, Menoufiya University, Egypt
| | | | | | | |
Collapse
|
45
|
Luo Y, Liu J, Wang Y, Su J, Wu Y, Hu G, Gao M, Quan F, Zhang Y. PhiC31 integrase-mediated genomic integration and stable gene expression in the mouse mammary gland after gene electrotransfer. J Gene Med 2013; 15:356-65. [PMID: 24288809 DOI: 10.1002/jgm.2723] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND PhiC31 integrase is capable of conferring long-term transgene expression in various transfected tissues in vivo. In the present study, we investigated the activity of phiC31 integrase in mouse mammary glands. METHODS The normal mouse mammary epithelial cell line HC11 was transfected with FuGENE® HD Transfection Reagent (Roche Diagnostics, Shanghai, China). Transfection of the mouse mammary gland in vivo was performed by electrotransfer. Transgene expression was detected by western blotting and an enzyme-linked immunosorbent assay. Genomic integration and integration at mpsL1 was confirmed by a nested polymerase chain reaction. RESULTS An optimal electrotransfer protocol for the lactating mouse mammary gland was attained through investigation of different voltages and pulse durations. PhiC31 integrase mediated site-specific transgene integration in HC11 cells and the mouse mammary gland. In addition, the site-specific integration occurred efficiently at the ‘hot spot’ mpsL1. Co-delivery of PhiC31 integrase enhanced and prolonged transgene expression in the mouse mammary gland. CONCLUSIONS The results obtained in the present study show that the use of phiC31 integrase is a feasible and efficient method for high and stable transgene expression in the mouse mammary gland.
Collapse
|
46
|
Gene therapy and DNA delivery systems. Int J Pharm 2013; 459:70-83. [PMID: 24286924 DOI: 10.1016/j.ijpharm.2013.11.041] [Citation(s) in RCA: 294] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 10/31/2013] [Accepted: 11/19/2013] [Indexed: 12/29/2022]
Abstract
Gene therapy is a promising new technique for treating many serious incurable diseases, such as cancer and genetic disorders. The main problem limiting the application of this strategy in vivo is the difficulty of transporting large, fragile and negatively charged molecules like DNA into the nucleus of the cell without degradation. The key to success of gene therapy is to create safe and efficient gene delivery vehicles. Ideally, the vehicle must be able to remain in the bloodstream for a long time and avoid uptake by the mononuclear phagocyte system, in order to ensure its arrival at the desired targets. Moreover, this carrier must also be able to transport the DNA efficiently into the cell cytoplasm, avoiding lysosomal degradation. Viral vehicles are the most commonly used carriers for delivering DNA and have long been used for their high efficiency. However, these vehicles can trigger dangerous immunological responses. Scientists need to find safer and cheaper alternatives. Consequently, the non-viral carriers are being prepared and developed until techniques for encapsulating DNA can be found. This review highlights gene therapy as a new promising technique used to treat many incurable diseases and the different strategies used to transfer DNA, taking into account that introducing DNA into the cell nucleus without degradation is essential for the success of this therapeutic technique.
Collapse
|
47
|
Kariya S, Komemushi A, Nakatani M, Yoshida R, Sawada S, Tanigawa N. Re: CO2 microbubble contrast enhancement in x-ray angiography. A reply. Clin Radiol 2013; 68:1180. [DOI: 10.1016/j.crad.2013.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 06/03/2013] [Indexed: 11/26/2022]
|
48
|
Chen B, Guo F, Li G, Wang P. Three-Dimensional Simulation of Bubble Formation Through a Microchannel T-Junction. Chem Eng Technol 2013. [DOI: 10.1002/ceat.201300169] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
49
|
Targeted gene transfer into head and neck squamous cell carcinoma by nanosecond pulsed laser-induced stress waves. Lasers Med Sci 2013; 29:231-8. [DOI: 10.1007/s10103-013-1324-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 04/15/2013] [Indexed: 11/25/2022]
|
50
|
Delalande A, Kotopoulis S, Postema M, Midoux P, Pichon C. Sonoporation: mechanistic insights and ongoing challenges for gene transfer. Gene 2013; 525:191-9. [PMID: 23566843 DOI: 10.1016/j.gene.2013.03.095] [Citation(s) in RCA: 143] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 02/27/2013] [Accepted: 03/07/2013] [Indexed: 11/29/2022]
Abstract
Microbubbles first developed as ultrasound contrast agents have been used to assist ultrasound for cellular drug and gene delivery. Their oscillation behavior during ultrasound exposure leads to transient membrane permeability of surrounding cells, facilitating targeted local delivery. The increased cell uptake of extracellular compounds by ultrasound in the presence of microbubbles is attributed to a phenomenon called sonoporation. In this review, we summarize current state of the art concerning microbubble-cell interactions and cellular effects leading to sonoporation and its application for gene delivery. Optimization of sonoporation protocol and composition of microbubbles for gene delivery are discussed.
Collapse
|