51
|
Cao WJ, Rosenblat JD, Roth NC, Kuliszewski MA, Matkar PN, Rudenko D, Liao C, Lee PJH, Leong-Poi H. Therapeutic Angiogenesis by Ultrasound-Mediated MicroRNA-126-3p Delivery. Arterioscler Thromb Vasc Biol 2015; 35:2401-11. [PMID: 26381870 DOI: 10.1161/atvbaha.115.306506] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 09/02/2015] [Indexed: 01/15/2023]
Abstract
OBJECTIVE MicroRNAs are involved in many critical functions, including angiogenesis. Ultrasound-targeted microbubble destruction (UTMD) is a noninvasive technique for targeted vascular transfection of plasmid DNA and may be well suited for proangiogenic microRNA delivery. We aimed to investigate UTMD of miR-126-3p for therapeutic angiogenesis in chronic ischemia. APPROACH AND RESULTS The angiogenic potential of miR-126-3p was tested in human umbilical vein endothelial cells in vitro. UTMD of miR-126-3p was tested in vivo in Fischer-344 rats before and after chronic left femoral artery ligation, evaluating target knockdown, miR-126-3p and miR-126-5p expression, phosphorylated Tie2 levels, microvascular perfusion, and vessel density. In vitro, miR-126-3p-transfected human umbilical vein endothelial cells showed repression of sprouty-related protein-1 and phosphatidylinositol-3-kinase regulatory subunit 2, negative regulators of vascular endothelial growth factor and angiopoietin-1 signaling, increased phosphorylated Tie2 mediated by knockdown of phosphatidylinositol-3-kinase regulatory subunit 2 and greater angiogenic potential mediated by both vascular endothelial growth factor/vascular endothelial growth factor R2 and angiopoietin-1 /Tie2 effects. UTMD of miR-126-3p resulted in targeted vascular transfection, peaking early after delivery and lasting for >3 days, and resulting in inhibition of sprouty-related protein-1 and phosphatidylinositol-3-kinase regulatory subunit 2, with minimal uptake in remote organs. Finally, UTMD of miR-126-3p to chronic ischemic hindlimb muscle resulted in improved perfusion, vessel density, enhanced arteriolar formation, pericyte coverage, and phosphorylated Tie2 levels, without affecting miR-126-5p or delta-like 1 homolog levels. CONCLUSIONS UTMD of miR-126 results in improved tissue perfusion and vascular density in the setting of chronic ischemia by repressing sprouty-related protein-1 and phosphatidylinositol-3-kinase regulatory subunit 2 and enhancing vascular endothelial growth factor and angiopoietin-1 signaling, with no effect on miR-126-5p. UTMD is a promising platform for microRNA delivery, with applications for therapeutic angiogenesis.
Collapse
Affiliation(s)
- Wei J Cao
- From the Division of Cardiology, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St Michael's Hospital, University of Toronto, Ontario, Canada
| | - Joshua D Rosenblat
- From the Division of Cardiology, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St Michael's Hospital, University of Toronto, Ontario, Canada
| | - Nathan C Roth
- From the Division of Cardiology, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St Michael's Hospital, University of Toronto, Ontario, Canada
| | - Michael A Kuliszewski
- From the Division of Cardiology, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St Michael's Hospital, University of Toronto, Ontario, Canada
| | - Pratiek N Matkar
- From the Division of Cardiology, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St Michael's Hospital, University of Toronto, Ontario, Canada
| | - Dmitriy Rudenko
- From the Division of Cardiology, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St Michael's Hospital, University of Toronto, Ontario, Canada
| | - Christine Liao
- From the Division of Cardiology, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St Michael's Hospital, University of Toronto, Ontario, Canada
| | - Paul J H Lee
- From the Division of Cardiology, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St Michael's Hospital, University of Toronto, Ontario, Canada
| | - Howard Leong-Poi
- From the Division of Cardiology, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St Michael's Hospital, University of Toronto, Ontario, Canada.
| |
Collapse
|
52
|
Abstract
Microbubbles are an excellent intravascular tracer, and both the rate of myocardial opacification (analogous to coronary microvascular perfusion) and contrast intensity (analogous to myocardial blood volume) provide unique insights into myocardial perfusion. A strong evidence base has been accumulated to show comparability with nuclear perfusion imaging and incremental diagnostic and prognostic value relative to wall motion analysis. This technique also provides the possibility to measure myocardial perfusion at the bedside. Despite all of these advantages, the technique is complicated, technically challenging, and has failed to scale legislative and financial hurdles. The development of targeted imaging and therapeutic interventions will hopefully rekindle interest in this interesting modality.
Collapse
Affiliation(s)
- Faraz Pathan
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Thomas H Marwick
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia.
| |
Collapse
|
53
|
Chen S, Chen J, Huang P, Meng XL, Clayton S, Shen JS, Grayburn PA. Myocardial regeneration in adriamycin cardiomyopathy by nuclear expression of GLP1 using ultrasound targeted microbubble destruction. Biochem Biophys Res Commun 2015; 458:823-9. [DOI: 10.1016/j.bbrc.2015.02.038] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 02/06/2015] [Indexed: 01/08/2023]
|
54
|
Ultrasound-guided delivery of microRNA loaded nanoparticles into cancer. J Control Release 2015; 203:99-108. [PMID: 25687306 DOI: 10.1016/j.jconrel.2015.02.018] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 02/04/2015] [Accepted: 02/13/2015] [Indexed: 11/20/2022]
Abstract
Ultrasound induced microbubble cavitation can cause enhanced permeability across natural barriers of tumors such as vessel walls or cellular membranes, allowing for enhanced therapeutic delivery into the target tissues. While enhanced delivery of small (<1nm) molecules has been shown at acoustic pressures below 1MPa both in vitro and in vivo, the delivery efficiency of larger (>100nm) therapeutic carriers into cancer remains unclear and may require a higher pressure for sufficient delivery. Enhanced delivery of larger therapeutic carriers such as FDA approved pegylated poly(lactic-co-glycolic acid) nanoparticles (PLGA-PEG-NP) has significant clinical value because these nanoparticles have been shown to protect encapsulated drugs from degradation in the blood circulation and allow for slow and prolonged release of encapsulated drugs at the target location. In this study, various acoustic parameters were investigated to facilitate the successful delivery of two nanocarriers, a fluorescent semiconducting polymer model drug nanoparticle as well as PLGA-PEG-NP into human colon cancer xenografts in mice. We first measured the cavitation dose produced by various acoustic parameters (pressure, pulse length, and pulse repetition frequency) and microbubble concentration in a tissue mimicking phantom. Next, in vivo studies were performed to evaluate the penetration depth of nanocarriers using various acoustic pressures, ranging between 1.7 and 6.9MPa. Finally, a therapeutic microRNA, miR-122, was loaded into PLGA-PEG-NP and the amount of delivered miR-122 was assessed using quantitative RT-PCR. Our results show that acoustic pressures had the strongest effect on cavitation. An increase of the pressure from 0.8 to 6.9MPa resulted in a nearly 50-fold increase in cavitation in phantom experiments. In vivo, as the pressures increased from 1.7 to 6.9MPa, the amount of nanoparticles deposited in cancer xenografts was increased from 4- to 14-fold, and the median penetration depth of extravasated nanoparticles was increased from 1.3-fold to 3-fold, compared to control conditions without ultrasound, as examined on 3D confocal microscopy. When delivering miR-122 loaded PLGA-PEG-NP using optimal acoustic settings with minimum tissue damage, miR-122 delivery into tumors with ultrasound and microbubbles was 7.9-fold higher compared to treatment without ultrasound. This study demonstrates that ultrasound induced microbubble cavitation can be a useful tool for delivery of therapeutic miR loaded nanocarriers into cancer in vivo.
Collapse
|
55
|
Sanches PG, Mühlmeister M, Seip R, Kaijzel E, Löwik C, Böhmer M, Tiemann K, Grüll H. Ultrasound-mediated gene delivery of naked plasmid DNA in skeletal muscles: A case for bolus injections. J Control Release 2014; 195:130-7. [DOI: 10.1016/j.jconrel.2014.06.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 06/06/2014] [Accepted: 06/20/2014] [Indexed: 12/17/2022]
|
56
|
Yan L, Zhang J, Lee CS, Chen X. Micro- and nanotechnologies for intracellular delivery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2014; 10:4487-504. [PMID: 25168360 DOI: 10.1002/smll.201401532] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 07/18/2014] [Indexed: 05/24/2023]
Abstract
The majority of drugs and biomolecules need to be delivered into cells to be effective. However, the cell membranes, a biological barrier, strictly resist drugs or biomolecules entering cells, resulting in significantly reduced intracellular delivery efficiency. To overcome this barrier, a variety of intracellular delivery approaches including chemical and physical ways have been developed in recent years. In this review, the focus is on summarizing the nanomaterial routes involved in making use of a collection of receptors for the targeted delivery of drugs and biomolecules and the physical ways of applying micro- and nanotechnologies for high-throughput intracellular delivery.
Collapse
Affiliation(s)
- Li Yan
- Center of Super-Diamond and Advanced Films (COSDAF) and Department of Physics and Materials Science, City University of Hong Kong, Hong Kong SAR, PR China
| | | | | | | |
Collapse
|
57
|
Gao R, Zhou X, Yang Y, Wang Z. Transfection of wtp53 and Rb94 genes into retinoblastomas of nude mice by ultrasound-targeted microbubble destruction. ULTRASOUND IN MEDICINE & BIOLOGY 2014; 40:2662-2670. [PMID: 25218456 DOI: 10.1016/j.ultrasmedbio.2014.05.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 05/09/2014] [Accepted: 05/19/2014] [Indexed: 06/03/2023]
Abstract
Using ultrasound-targeted microbubble destruction (UTMD), we transfected both wild-type p53 (wtp53) and Rb94 genes into retinoblastomas (RBs) of nude mice to investigate the inhibitory role of these two genes in RB development. The 40 tumor-bearing mice, which had been established by sub-retinal injection of an HXO-Rb44 cell suspension, were randomly divided into five groups: blank control group (C); blank plasmid group (M); wtp53 plasmid group (p53); Rb94 plasmid group (Rb94); wtp53 + Rb94 plasmid group (p53 + Rb94). For preparation of the DNA-loaded microbubbles, a pre-determined amount of blank plasmid, pVIVO1-p53, pVIVO1-Rb94 or pVIVO1-p53-Rb94 was added and mixed with the microbubbles. Then, these DNA-loaded microbubbles were respectively transfected into the animal model by UTMD. Vascular endothelial growth factor level and microvessel density of the tumor were determined by immunohistochemical staining. Apoptosis of tissues was detected by terminal deoxynucleotidyl transferase dUTP nick end labeling staining. Expression of wtp53 and Rb94 at both the gene and protein levels was detected by RT-PCR (reverse transcription polymerase chain reaction) and Western blot, respectively. Transfection of both genes had greater inhibitory effects on RB development and resulted in lower levels of vascular endothelial growth factor, lower microvessel density and more obvious apoptosis than single-gene transfection (p < 0.05). The results indicate that the transfection of both genes into the RB by UTMD more strongly inhibited RB growth than transfection of a single gene.
Collapse
Affiliation(s)
- Ruiqi Gao
- Department of Ophthalmology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China; Department of Ophthalmology, Dujiangyan Medical Center, Sichuan, China
| | - Xiyuan Zhou
- Department of Ophthalmology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Yingxue Yang
- Department of Ophthalmology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhigang Wang
- Institute of Ultrasonic Imaging, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Ophthalmology, Chongqing, China
| |
Collapse
|
58
|
Borrajo E, Vidal A, Alonso MJ, Garcia‐Fuentes M. How Regenerative Medicine Can Benefit from Nucleic Acids Delivery Nanocarriers? POLYMERS IN REGENERATIVE MEDICINE 2014:285-336. [DOI: 10.1002/9781118356692.ch9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
|
59
|
High-frequency ultrasound-guided disruption of glycoprotein VI-targeted microbubbles targets atheroprogressison in mice. Biomaterials 2014; 36:80-9. [PMID: 25301636 DOI: 10.1016/j.biomaterials.2014.09.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 09/16/2014] [Indexed: 01/25/2023]
Abstract
Targeted contrast-enhanced ultrasound (CEU) using microbubble agents is a promising non-invasive imaging technique to evaluate atherosclerotic lesions. In this study, we decipher the diagnostic and therapeutic potential of targeted-CEU with soluble glycoprotein (GP)-VI in vivo. Microbubbles were conjugated with the recombinant fusion protein GPVI-Fc (MBGPVI) that binds with high affinity to atherosclerotic lesions. MBGPVI or control microbubbles (MBC) were intravenously administered into ApoE(-/-) or wild type mice and binding of the microbubbles to the vessel wall was visualized by high-resolution CEU. CEU molecular imaging signals of MBGPVI were substantially enhanced in the aortic arch and in the truncus brachiocephalicus in ApoE(-/-) as compared to wild type mice. High-frequency ultrasound (HFU)-guided disruption of MBGPVI enhanced accumulation of GPVI in the atherosclerotic lesions, which may interfere with atheroprogression. Thus, we establish targeted-CEU with soluble GPVI as a novel non-invasive molecular imaging method for atherosclerosis. Further, HFU-guided disruption of GPVI-targeted microbubbles is an innovate therapeutic approach that potentially prevents progression of atherosclerotic disease.
Collapse
|
60
|
Xia M, Conley SM, Li G, Li PL, Boini KM. Inhibition of hyperhomocysteinemia-induced inflammasome activation and glomerular sclerosis by NLRP3 gene deletion. Cell Physiol Biochem 2014; 34:829-41. [PMID: 25171193 DOI: 10.1159/000363046] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2014] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND/AIMS Hyperhomocysteinemia (hHcys) has been reported to initiate Nod-like receptor protein 3 (NLRP3) inflammasome formation and activation in podocytes, leading to glomerular dysfunction and sclerosis. However, it remains unknown whether Nlrp3 gene is critical for the formation and activation of inflammasomes in glomeruli of hHcys mice. METHODS Plasma homocysteine concentration was estimated utilizing HPLC, inflammasome formation and immunofluorescence expression from confocal microscopy, IL-1β production from ELISA. RESULTS Uninephrectomized Nlrp3 knockout (Nlrp3(-/-)) and wild type (Nlrp3(+/+)) and intra renal Nlrp3 shRNA-transfected wild type mice (Nlrp3 shRNA) were fed a folate free (FF) diet or normal chow (ND) for 4 weeks to produce hHcys. The plasma Hcys levels were significantly elevated in both Nlrp3(-/-) and Nlrp3(+/+) mice fed a FF diet compared to ND fed mice. The FF diet significantly increased the colocalization of Nlrp3 with apoptosis-associated speck-like protein (ASC) or caspase-1, caspase-1 activity and IL-1β production in glomeruli of Nlrp3(+/+), but not in Nlrp3(-/-) mice and local Nlrp3 shRNA transfected mice. Correspondingly, the glomerular damage index (GDI) and urinary protein excretion were significantly higher in Nlrp3(+/+) mice compared to ND fed mice. However, the hHcys-induced increase in GDI and proteinuria were significantly lower in Nlrp3(-/-) and local Nlrp3 shRNA transfected mice than in Nlrp3(+/+) mice. Immunocytochemical analysis showed that hHcys decreased expression of podocin and nephrin, but increased desmin expression in glomeruli of Nlrp3(+/+) mice compared to Nlrp3(-/-) mice. CONCLUSION Nlrp3 gene is an essential component of Nlrp3 inflammasomes and that targeting Nlrp3 may be important therapeutic strategy to prevent inflammasome activation and thereby protect podocytes and glomeruli from hHcys-induced injury.
Collapse
Affiliation(s)
- Min Xia
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | | | | | | | | |
Collapse
|
61
|
Katz MG, Fargnoli AS, Williams RD, Bridges CR. Gene therapy delivery systems for enhancing viral and nonviral vectors for cardiac diseases: current concepts and future applications. Hum Gene Ther 2014; 24:914-27. [PMID: 24164239 DOI: 10.1089/hum.2013.2517] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Gene therapy is one of the most promising fields for developing new treatments for the advanced stages of ischemic and monogenetic, particularly autosomal or X-linked recessive, cardiomyopathies. The remarkable ongoing efforts in advancing various targets have largely been inspired by the results that have been achieved in several notable gene therapy trials, such as the hemophilia B and Leber's congenital amaurosis. Rate-limiting problems preventing successful clinical application in the cardiac disease area, however, are primarily attributable to inefficient gene transfer, host responses, and the lack of sustainable therapeutic transgene expression. It is arguable that these problems are directly correlated with the choice of vector, dose level, and associated cardiac delivery approach as a whole treatment system. Essentially, a delicate balance exists in maximizing gene transfer required for efficacy while remaining within safety limits. Therefore, the development of safe, effective, and clinically applicable gene delivery techniques for selected nonviral and viral vectors will certainly be invaluable in obtaining future regulatory approvals. The choice of gene transfer vector, dose level, and the delivery system are likely to be critical determinants of therapeutic efficacy. It is here that the interactions between vector uptake and trafficking, delivery route means, and the host's physical limits must be considered synergistically for a successful treatment course.
Collapse
Affiliation(s)
- Michael G Katz
- Sanger Heart and Vascular Institute , Cannon Research Center, Carolinas HealthCare System, Charlotte, NC 28203
| | | | | | | |
Collapse
|
62
|
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: 8.3] [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
|
63
|
Reduced Ischemic Injury After Stroke in Mice by Angiogenic Gene Delivery Via Ultrasound-Targeted Microbubble Destruction. J Neuropathol Exp Neurol 2014; 73:548-58. [DOI: 10.1097/nen.0000000000000077] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
64
|
Kooiman K, Vos HJ, Versluis M, de Jong N. Acoustic behavior of microbubbles and implications for drug delivery. Adv Drug Deliv Rev 2014; 72:28-48. [PMID: 24667643 DOI: 10.1016/j.addr.2014.03.003] [Citation(s) in RCA: 249] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 02/11/2014] [Accepted: 03/18/2014] [Indexed: 12/21/2022]
Abstract
Ultrasound contrast agents are valuable in diagnostic ultrasound imaging, and they increasingly show potential for drug delivery. This review focuses on the acoustic behavior of flexible-coated microbubbles and rigid-coated microcapsules and their contribution to enhanced drug delivery. Phenomena relevant to drug delivery, such as non-spherical oscillations, shear stress, microstreaming, and jetting will be reviewed from both a theoretical and experimental perspective. Further, the two systems for drug delivery, co-administration and the microbubble as drug carrier system, are reviewed in relation to the microbubble behavior. Finally, future prospects are discussed that need to be addressed for ultrasound contrast agents to move from a pre-clinical tool into a clinical setting.
Collapse
|
65
|
New progress in angiogenesis therapy of cardiovascular disease by ultrasound targeted microbubble destruction. BIOMED RESEARCH INTERNATIONAL 2014; 2014:872984. [PMID: 24900995 PMCID: PMC4037580 DOI: 10.1155/2014/872984] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 03/26/2014] [Indexed: 02/08/2023]
Abstract
Angiogenesis plays a vital part in the pathogenesis and treatment of cardiovascular disease and has become one of the hotspots that are being discussed in the past decades. At present, the promising angiogenesis therapies are gene therapy and stem cell therapy. Besides, a series of studies have shown that the ultrasound targeted microbubble destruction (UTMD) was a novel gene delivery system, due to its advantages of noninvasiveness, low immunogenicity and toxicity, repeatability and temporal and spatial target specificity; UTMD has also been used for angiogenesis therapy of cardiovascular disease. In this review, we mainly discuss the combination of UTMD and gene therapy or stem cell therapy which is applied in angiogenesis therapy in recent researches, and outline the future challenges and good prospects of these approaches.
Collapse
|
66
|
Guo C, Jin Y, Dai Z. Multifunctional Ultrasound Contrast Agents for Imaging Guided Photothermal Therapy. Bioconjug Chem 2014; 25:840-54. [DOI: 10.1021/bc500092h] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Caixin Guo
- School
of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Yushen Jin
- School
of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
- Department
of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, China
| | - Zhifei Dai
- Department
of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, China
| |
Collapse
|
67
|
Huang L, Belousova T, Pan JSC, Du J, Ju H, Lu L, Zhang P, Truong LD, Nuotio-Antar A, Sheikh-Hamad D. AKI after conditional and kidney-specific knockdown of stanniocalcin-1. J Am Soc Nephrol 2014; 25:2303-15. [PMID: 24700878 DOI: 10.1681/asn.2013070690] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Stanniocalcin-1 is an intracrine protein; it binds to the cell surface, is internalized to the mitochondria, and diminishes superoxide generation through induction of uncoupling proteins. In vitro, stanniocalcin-1 inhibits macrophages and preserves endothelial barrier function, and transgenic overexpression of stanniocalcin-1 in mice protects against ischemia-reperfusion kidney injury. We sought to determine the kidney phenotype after kidney endothelium-specific expression of stanniocalcin-1 small hairpin RNA (shRNA). We generated transgenic mice that express stanniocalcin-1 shRNA or scrambled shRNA upon removal of a floxed reporter (phosphoglycerate kinase-driven enhanced green fluorescent protein) and used ultrasound microbubbles to deliver tyrosine kinase receptor-2 promoter-driven Cre to the kidney to permit kidney endothelium-specific shRNA expression. Stanniocalcin-1 mRNA and protein were expressed throughout the kidney in wild-type mice. Delivery of tyrosine kinase receptor-2 promoter-driven Cre to stanniocalcin-1 shRNA transgenic kidneys diminished the expression of stanniocalcin-1 mRNA and protein throughout the kidneys. Stanniocalcin-1 mRNA and protein expression did not change in similarly treated scrambled shRNA transgenic kidneys, and we observed no Cre protein expression in cultured and tyrosine kinase receptor-2 promoter-driven Cre-transfected proximal tubule cells, suggesting that knockdown of stanniocalcin-1 in epithelial cells in vivo may result from stanniocalcin-1 shRNA transfer from endothelial cells to epithelial cells. Kidney-specific knockdown of stanniocalcin-1 led to severe proximal tubule injury characterized by vacuolization, decreased uncoupling of protein-2 expression, greater generation of superoxide, activation of the unfolded protein response, initiation of autophagy, cell apoptosis, and kidney failure. Our observations suggest that stanniocalcin-1 is critical for tubular epithelial survival under physiologic conditions.
Collapse
Affiliation(s)
| | | | | | - Jie Du
- Department of Medicine/Division of Nephrology
| | - Huiming Ju
- Department of Medicine/Division of Nephrology
| | - Lianghao Lu
- Department of Medicine/Division of Nephrology
| | - Pumin Zhang
- Department of Molecular Physiology and Biophysics, and
| | - Luan D Truong
- Kidney Pathology Laboratory, The Methodist Hospital/Weill Cornell University, Houston, Texas
| | - Alli Nuotio-Antar
- Department of Pediatrics/Nutrition, Baylor College of Medicine, Houston, Texas; and
| | | |
Collapse
|
68
|
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: 1.4] [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
|
69
|
Xia M, Boini KM, Abais JM, Xu M, Zhang Y, Li PL. Endothelial NLRP3 inflammasome activation and enhanced neointima formation in mice by adipokine visfatin. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:1617-28. [PMID: 24631027 DOI: 10.1016/j.ajpath.2014.01.032] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 01/22/2014] [Accepted: 01/30/2014] [Indexed: 12/25/2022]
Abstract
Inflammasomes serve as an intracellular machinery to initiate inflammatory response to various danger signals. The present study tested whether an inflammasome centered on nucleotide oligomerization domain-like receptor protein 3 (NLRP3) triggers endothelial inflammatory response to adipokine visfatin, a major injurious adipokine during obesity. NLRP3 inflammasome components were abundantly expressed in cultured mouse microvascular endothelial cells, including NLRP3, apoptosis-associated speck-like protein, and caspase-1. These NLRP3 inflammasome molecules could be aggregated to form an inflammasome complex on stimulation of visfatin, as shown by fluorescence confocal microscopy and size exclusion chromatography. Correspondingly, visfatin significantly increased caspase-1 activity and IL-1β release in microvascular endothelial cells, indicating an activation of NLRP3 inflammasomes. In animal experiments, direct infusion of visfatin in mice with partially ligated left carotid artery were found to have significantly increased neointimal formation, which was correlated with increased NLRP3 inflammasome formation and IL-1β production in the intima. Further, visfatin-induced neointimal formation, endothelial inflammasome formation, and IL-1β production in mouse partially ligated left carotid artery were abolished by caspase-1 inhibition, local delivery of apoptosis-associated speck-like protein shRNA or deletion of the ASC gene. In conclusion, the formation and activation of NLRP3 inflammasomes by adipokine visfatin may be an important initiating mechanism to turn on the endothelial inflammatory response leading to arterial inflammation and endothelial dysfunction in mice during early stage obesity.
Collapse
Affiliation(s)
- Min Xia
- Department of Pharmacology & Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond, Virginia
| | - Krishna M Boini
- Department of Pharmacology & Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond, Virginia
| | - Justine M Abais
- Department of Pharmacology & Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond, Virginia
| | - Ming Xu
- Department of Pharmacology & Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond, Virginia
| | - Yang Zhang
- Department of Pharmacology & Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond, Virginia
| | - Pin-Lan Li
- Department of Pharmacology & Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond, Virginia.
| |
Collapse
|
70
|
Kilroy JP, Patil AV, Rychak JJ, Hossack JA. An IVUS transducer for microbubble therapies. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2014; 61:441-9. [PMID: 24569249 PMCID: PMC4136497 DOI: 10.1109/tuffc.2014.2929] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
There is interest in examining the potential of modified intravascular ultrasound (IVUS) catheters to facilitate dual diagnostic and therapeutic roles using ultrasound plus microbubbles for localized drug delivery to the vessel wall. The goal of this study was to design, prototype, and validate an IVUS transducer for microbubble-based drug delivery. A 1-D acoustic radiation force model and finite element analysis guided the design of a 1.5-MHz IVUS transducer. Using the IVUS transducer, biotinylated microbubbles were displaced in water and bovine whole blood to the streptavidin-coated wall of a flow phantom by a 1.5-MHz center frequency, peak negative pressure = 70 kPa pulse with varying pulse repetition frequency (PRF) while monitoring microbubble adhesion with ultrasound. A fit was applied to the RF data to extract a time constant (τ). As PRF was increased in water, the time constant decreased (τ = 32.6 s, 1 kHz vs. τ = 8.2 s, 6 kHz), whereas in bovine whole blood an adhesion-no adhesion transition was found for PRFs ≥ 8 kHz. Finally, a fluorophore was delivered to an ex vivo swine artery using microbubbles and the IVUS transducer, resulting in a 6.6-fold increase in fluorescence. These results indicate the importance of PRF (or duty factor) for IVUS acoustic radiation force microbubble displacement and the potential for IVUS and microbubbles to provide localized drug delivery.
Collapse
Affiliation(s)
- Joseph P. Kilroy
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| | | | - Joshua J. Rychak
- Targeson Inc. and the Department of Bioengineering, University of California, San Diego, CA
| | - John A. Hossack
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| |
Collapse
|
71
|
Zhang Y, Huang XR, Wei LH, Chung AC, Yu CM, Lan HY. miR-29b as a therapeutic agent for angiotensin II-induced cardiac fibrosis by targeting TGF-β/Smad3 signaling. Mol Ther 2014; 22:974-85. [PMID: 24569834 DOI: 10.1038/mt.2014.25] [Citation(s) in RCA: 234] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2013] [Accepted: 02/10/2014] [Indexed: 12/17/2022] Open
Abstract
Loss of miR-29 is associated with cardiac fibrosis. This study examined the role and therapeutic potential of miR-29 in mouse model of hypertension induced by angiotensin II (AngII). By using microRNA microarray, in situ hybridization, and real-time polymerase chain reaction, we found that AngII-induced cardiac fibrosis in the hypertensive heart and in cultured cardiac fibroblasts were associated with downregulation of miR-29a-c via a Smad3-dependent mechanism. In vitro knockdown of miR-29b enhanced but overexpression of miR-29b inhibited AngII-induced fibrosis, revealing a protective role of miR-29b in cardiac fibrosis in response to AngII. This was further demonstrated in vivo by the ability of overexpressing miR-29b in the mouse heart to prevent AngII-mediated cardiac fibrosis and cardiac dysfunction. Importantly, we also found that restored miR-29b in the established hypertensive heart was capable of blocking progressive cardiac fibrosis and improving cardiac dysfunction, demonstrating a therapeutic potential of miR-29b for chronic heart disease. Further studies revealed that targeting the transforming growth factor (TGF)-β1 coding sequence region, thereby inhibiting TGF-β/Smad3 signaling, could be a new mechanism by which miR-29b inhibited AngII-induced cardiac fibrosis. In conclusion, miR-29b plays a protective role in AngII-mediated cardiac remodeling and may be a therapeutic agent for cardiac fibrosis by targeting the TGF-β/Smad3 pathway.
Collapse
Affiliation(s)
- Yang Zhang
- 1] Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China [2] Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiao-Ru Huang
- 1] Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China [2] Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Li-Hua Wei
- 1] Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China [2] Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Arthur Ck Chung
- 1] Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China [2] Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Cheuk-Man Yu
- 1] Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China [2] Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Hui-Yao Lan
- 1] Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China [2] Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
72
|
The road ahead: working towards effective clinical translation of myocardial gene therapies. Ther Deliv 2014; 5:39-51. [PMID: 24341816 DOI: 10.4155/tde.13.134] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
During the last two decades the fields of molecular and cellular cardiology, and more recently molecular cardiac surgery, have developed rapidly. The concept of delivering cDNA encoding a therapeutic gene to cardiomyocytes using a vector system with substantial cardiac tropism, allowing for long-term expression of a therapeutic protein, has moved from hypothesis to bench to clinical application. However, the clinical results to date are still disappointing. The ideal gene transfer method should be explored in clinically relevant animal models of heart disease to evaluate the relative roles of specific molecular pathways in disease pathogenesis, helping to validate the potential targets for therapeutic intervention. Successful clinical cardiovascular gene therapy also requires the use of nonimmunogenic cardiotropic vectors capable of expressing the requisite amount of therapeutic protein in vivo and in situ. Depending on the desired application either regional or global myocardial gene delivery is required. Cardiac-specific delivery techniques incorporating mapping technologies for regional delivery and highly efficient methodologies for global delivery should improve the precision and specificity of gene transfer to the areas of interest and minimize collateral organ gene expression.
Collapse
|
73
|
Bukhari F, MacGillivray T, del Monte F, Hajjar RJ. Genetic maneuvers to ameliorate ventricular function in heart failure: therapeutic potential and future implications. Expert Rev Cardiovasc Ther 2014; 3:85-97. [PMID: 15723577 DOI: 10.1586/14779072.3.1.85] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Gene therapy to treat heart failure has evolved into a growing field of investigation yielding remarkable results in preclinical models. Whether these results will persist in clinical trials remains to be seen. However, researchers still face a number of obstacles that need to be overcome before this treatment can be employed effectively. Efforts are required to identify better vectors with minimal side effects and maximal efficiency and durability. There is also a need to develop less invasive and more effective techniques to deliver these vectors. This review will discuss different methods to achieve these goals, the various pathologic mechanisms that have been targeted so far and those with strong potential for use in the future.
Collapse
Affiliation(s)
- Fariya Bukhari
- University of Arizona, Department of Medicine, Tucson, AZ 85721, USA.
| | | | | | | |
Collapse
|
74
|
Lee PJH, Rudenko D, Kuliszewski MA, Liao C, Kabir MG, Connelly KA, Leong-Poi H. Survivin gene therapy attenuates left ventricular systolic dysfunction in doxorubicin cardiomyopathy by reducing apoptosis and fibrosis. Cardiovasc Res 2014; 101:423-33. [PMID: 24403316 DOI: 10.1093/cvr/cvu001] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
AIMS The aim of this study was to investigate anti-apoptotic gene therapy using ultrasound-mediated plasmid delivery of survivin, an inhibitor of apoptosis protein, to prevent apoptosis and to attenuate left ventricular (LV) systolic dysfunction in a model of heart failure induced by doxorubicin. METHODS AND RESULTS Effect of survivin transduction was investigated in vitro in rat cardiomyoblasts. After survivin transduction, survivin protein was detected in cell culture supernate confirming secretion of extracellular survivin. Under doxorubicin stimulation, survivin-transduced cells had significantly reduced apoptosis; however, incubation with survivin-conditioned media also showed reduced apoptosis that was absent with null-conditioned media. Doxorubicin-induced cardiomyopathy was established in Fischer rats. Subsets of animals underwent ultrasound-mediated survivin gene delivery or empty vector gene delivery at Week 3. Control rats received doxorubicin alone. Animals were studied using PCR, immunohistochemistry, echocardiography, and invasive haemodynamic studies out to Week 6. By Week 6, LV % fractional shortening by echocardiography and systolic function by pressure-volume loops were greater in survivin treated when compared with control- and empty-treated animals. There was reduced apoptosis by TUNEL and caspase activity in survivin-treated animals compared with control and empty treated at Week 4, with reduced interstitial fibrosis at Week 6. CONCLUSION Survivin gene therapy can attenuate the progression of LV systolic dysfunction in doxorubicin cardiomyopathy. This effect can be attributed to decreased myocyte apoptosis and prevention of maladaptive LV remodelling, by both direct myocyte transfection and potentially by paracrine mechanisms.
Collapse
Affiliation(s)
- Paul J H Lee
- Division of Cardiology, Keenan Research Centre in the Li Ka Shing Knowledge Institute, 6-044 Donnelly Wing, St Michael's Hospital, University of Toronto, 30 Bond Street, Toronto, ON, Canada M5B 1W8
| | | | | | | | | | | | | |
Collapse
|
75
|
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: 302] [Impact Index Per Article: 25.2] [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
|
76
|
Tong HP, Wang LF, Guo YL, Li L, Fan XZ, Ding J, Huang HY. Preparation of protamine cationic nanobubbles and experimental study of their physical properties and in vivo contrast enhancement. ULTRASOUND IN MEDICINE & BIOLOGY 2013; 39:2147-2157. [PMID: 23932278 DOI: 10.1016/j.ultrasmedbio.2013.05.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 05/17/2013] [Accepted: 05/24/2013] [Indexed: 06/02/2023]
Abstract
In this study, we aimed to prepare a novel type of microbubble (MB), protamine cationic nanobubble (NB), to provide a new vector for tumor gene therapy. We prepared cationic NBs with protamine and other lipid components using mechanical oscillation. The protamine cationic NBs had a mean diameter of 521.2 ± 37.57 nm, a zeta potential of +18.5 mV, and a gene-carrying capacity of 15.69 μg androgen receptor (AR) siRNA per 10(8) NBs. The cationic NBs exhibited superior contrast enhancement for in vivo imaging compared with SonoVue (Bracco, Geneva, Switzerland), and their physical properties did not change significantly after 1 wk; meanwhile, the transfection efficiency of the cationic NBs in androgen-independent prostate cancer cells mediated by ultrasound irradiation was better than that of liposomes (82.17 ± 7.4% vs. 55.04 ± 5.4%, p < 0.01). Therefore, the protamine cationic NB can be considered for use as a novel type of gene-loading MB for ultrasound imaging and MB-mediated gene therapy of tumors.
Collapse
Affiliation(s)
- Hai-Peng Tong
- Department of Ultrasound, Southwest Hospital, Third Military Medical University, Chongqing, China
| | | | | | | | | | | | | |
Collapse
|
77
|
A novel plasmid and SonoVue formulation plus ultrasound sonication for effective gene delivery in nude mice. Life Sci 2013; 93:536-42. [DOI: 10.1016/j.lfs.2013.08.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 07/13/2013] [Accepted: 08/19/2013] [Indexed: 12/21/2022]
|
78
|
Jin Q, Wang Z, Yan F, Deng Z, Ni F, Wu J, Shandas R, Liu X, Zheng H. A novel cationic microbubble coated with stearic acid-modified polyethylenimine to enhance DNA loading and gene delivery by ultrasound. PLoS One 2013; 8:e76544. [PMID: 24086748 PMCID: PMC3784428 DOI: 10.1371/journal.pone.0076544] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 08/27/2013] [Indexed: 11/18/2022] Open
Abstract
A novel cationic microbubble (MB) for improvement of the DNA loading capacity and the ultrasound-mediated gene delivery efficiency has been developed; it has been prepared with commercial lipids and a stearic acid modified polyethylenimine 600 (Stearic-PEI600) polymer synthesized via acylation reaction of branched PEI600 and stearic acid mediated by N, N'-carbonyldiimidazole (CDI). The MBs’ concentration, size distribution, stability and zeta potential (ζ-potential) were measured and the DNA loading capacity was examined as a function of the amount of Stearic-PEI600. The gene transfection efficiency and cytotoxicity were also examined using breast cancer MCF-7 cells via the reporter plasmid pCMV-Luc, encoding the firefly luciferase gene. The results showed that the Stearic-PEI600 polymer caused a significant increase in magnitude of ζ-potential of MBs. The addition of DNA into cationic MBs can shift ζ-potentials from positive to negative values. The DNA loading capacity of the MBs grew linearly from (5±0.2) ×10−3 pg/µm2 to (20±1.8) ×10−3 pg/µm2 when Stearic-PEI600 was increased from 5 mol% to 30 mol%. Transfection of MCF-7 cells using 5% PEI600 MBs plus ultrasound exposure yielded 5.76±2.58×103 p/s/cm2/sr average radiance intensity, was 8.97- and 7.53-fold higher than those treated with plain MBs plus ultrasound (6.41±5.82) ×102 p/s/cm2/sr, (P<0.01) and PEI600 MBs without ultrasound (7.65±6.18) ×102 p/s/cm2/sr, (P<0.01), respectively. However, the PEI600 MBs showed slightly higher cytotoxicity than plain MBs. The cells treated with PEI600-MBs and plain MBs plus ultrasound showed 59.5±6.1% and 71.4±7.1% cell viability, respectively. In conclusion, our study demonstrated that the novel cationic MBs were able to increase DNA loading capacity and gene transfection efficiency and could be potentially applied in targeted gene delivery and therapy.
Collapse
Affiliation(s)
- Qiaofeng Jin
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhiyong Wang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Fei Yan
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- * E-mail: (HZ); (FY)
| | - Zhiting Deng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Fei Ni
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Junru Wu
- Department of Physics, University of Vermont, Burlington, Vermont, United States of America
| | - Robin Shandas
- Department of Bioengineering, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Xin Liu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Key Lab for MRI, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Hairong Zheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- * E-mail: (HZ); (FY)
| |
Collapse
|
79
|
Yang D, Gao YH, Tan KB, Zuo ZX, Yang WX, Hua X, Li PJ, Zhang Y, Wang G. Inhibition of hepatic fibrosis with artificial microRNA using ultrasound and cationic liposome-bearing microbubbles. Gene Ther 2013; 20:1140-8. [PMID: 23966015 DOI: 10.1038/gt.2013.41] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 06/10/2013] [Accepted: 06/12/2013] [Indexed: 12/17/2022]
Abstract
We sought to investigate the antifibrotic effects of an artificial microRNA (miRNA) targeting connective tissue growth factor (CTGF) using the ultrasound-targeted cationic liposome-bearing microbubble destruction gene delivery system. Cationic liposomes were conjugated with microbubbles using a biotin-avidin system. Plasmids carrying the most effective artificial miRNA sequences were delivered by ultrasound-targeted cationic liposome-bearing microbubble destruction gene delivery system to rats with hepatic fibrosis. The results show that this method of gene delivery effectively transported the plasmids to the rat liver. The artificial miRNA reduced hepatic fibrosis pathological alterations as well as the protein and mRNA expressions of CTGF and transforming growth factor β1. Furthermore, the CTGF gene silencing decreased the levels of type I collagen and α-smooth muscle actin (P<0.01). These data suggest that delivery of an artificial miRNA targeted against CTGF using ultrasound-targeted cationic liposome-bearing microbubble destruction may be an efficacious therapeutic method to ameliorate hepatic fibrosis.
Collapse
Affiliation(s)
- D Yang
- 1] Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, China [2] Department of Ultrasound, 324th Military Hospital, Chongqing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Sugano M, Negishi Y, Endo-Takahashi Y, Hamano N, Usui M, Suzuki R, Maruyama K, Aramaki Y, Yamamoto M. Gene delivery to periodontal tissue using Bubble liposomes and ultrasound. J Periodontal Res 2013; 49:398-404. [PMID: 23889504 DOI: 10.1111/jre.12119] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/06/2013] [Indexed: 11/30/2022]
Abstract
BACKGROUND AND OBJECTIVE Periodontitis is the most common inflammatory disease caused by oral biofilm infection. For efficient periodontal treatment, it is important to enhance the outcome of existing regenerative therapies. The physical action of an ultrasound may be able to deliver a therapeutic gene or drugs into the local area of the periodontium being treated for periodontal regeneration. Previously, we developed "Bubble liposomes" as a useful carrier for gene or drug delivery, and reported that delivery efficiency was increased with high-frequency ultrasound in vitro and in vivo. Hence, the aim of the present study was to examine the possibility of delivering genes into gingival tissues using Bubble liposomes and ultrasound. MATERIAL AND METHODS We attempted to deliver naked plasmid DNA encoding luciferase or enhanced green fluorescent protein (EGFP) into the lower labial gingiva of Wistar rats using Bubble liposomes, with or without ultrasound exposure. Ultrasound parameters were optimized for intensity (0-4.0 W/cm(2) ) and exposure time (0-120 s) to establish the most efficient conditions for exposure. The efficacy and duration of gene expression in the gingiva were investigated using a luciferase assay and fluorescence microscopy. RESULTS The strongest relative luciferase activity was observed when rats were treated under the following ultrasound conditions: 2.0 W/cm(2) intensity and 30 s of exposure time. Relative luciferase activity, 1 d after gene delivery, was significantly higher in gingiva treated using Bubble liposomes and ultrasound than in gingiva of the other treatment groups. Histological analysis also showed that distinct EGFP-expressing cells were observed in transfected gingiva when rats were treated under optimized conditions. CONCLUSION From these results, the combination of Bubble liposomes and ultrasound provides an efficient technique for delivering plasmid DNA into the gingiva. This technique can be applied for the delivery of a variety of therapeutic molecules into target tissue, and may serve as a useful treatment strategy for periodontitis.
Collapse
Affiliation(s)
- M Sugano
- Department of Periodontology, Showa University School of Dentistry, Tokyo, Japan; Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Kidney-targeted transplantation of mesenchymal stem cells by ultrasound-targeted microbubble destruction promotes kidney repair in diabetic nephropathy rats. BIOMED RESEARCH INTERNATIONAL 2013; 2013:526367. [PMID: 23762850 PMCID: PMC3677660 DOI: 10.1155/2013/526367] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2013] [Revised: 04/19/2013] [Accepted: 04/19/2013] [Indexed: 01/02/2023]
Abstract
We test the hypothesis that ultrasound-targeted microbubble destruction (UTMD) technique increases the renoprotective effect of kidney-targeted transplantation of bone-marrow-derived mesenchymal stem cells (BM-MSCs) in diabetic nephropathy (DN) rats. Diabetes was induced by streptozotocin injection (60 mg/Kg, intraperitoneally) in Sprague-Dawley rats. MSCs were administered alone or in combination with UTMD to DN rats at 4 weeks after diabetes onset. Random blood glucose concentrations were measured at 1, 2, 4, and 8 weeks, and plasma insulin levels, urinary albumin excretion rate (UAER) values, the structures of pancreas and kidney, the expressions of TGF- β 1, synaptopodin, and IL-10 were assessed at 8 weeks after MSCs transplantation. MSCs transplantation decreased blood glucose concentrations and attenuated pancreatic islets/ β cells damage. The permeability of renal interstitial capillaries and VCAM-1 expression increased after UTMD, which enhanced homing and retention of MSCs to kidneys. MSCs transplantation together with UTMD prevented renal damage and decreased UAER values by inhibiting TGF- β 1 expression and upregulating synaptopodin and IL-10 expression. We conclude that MSCs transplantation reverts hyperglycemia; UTMD technique noninvasively increases the homing of MSCs to kidneys and promotes renal repair in DN rats. This noninvasive cell delivery method may be feasible and efficient as a novel approach for personal MSCs therapy to diabetic nephropathy.
Collapse
|
82
|
Ma J, DU LF, Chen M, Wang HH, Xing LX, Jing LF, Li YH. Drug-loaded nano-microcapsules delivery system mediated by ultrasound-targeted microbubble destruction: A promising therapy method. Biomed Rep 2013; 1:506-510. [PMID: 24648976 DOI: 10.3892/br.2013.110] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Accepted: 04/15/2013] [Indexed: 11/06/2022] Open
Abstract
The nano-microcapsules drug delivery system is currently a promising method for the treatment of many types of diseases, particularly tumors. However, the drug delivery efficiency does not reach a satisfactory level to meet treatment demands. Therefore, the effectiveness of delivery needs to be improved. Based on the alterations in the structure and modification of nano-microcapsules, ultrasound-targeted microbubble destruction (UTMD), a safe physical targeted method, may increase tissue penetration and cell membrane permeability, aiding the drug-loaded nano-microcapsules ingress the interior of targeted tissues and cells. The effectiveness and exact mechanism of action of the drug-loaded nano-microcapsules delivery system mediated by UTMD have yet to be fully elucidated. In this study, the latest advancement in UTMD-mediated drug loaded nano-microcapsules system technology was reviewed and the hindrances of UTMD-mediated drug delivery were assessed, in combination with a prospective study. The findings suggested that the drug delivery efficiency of nano-microcapsules mediated by UTMD was distinctly improved. Thus, the UTMD-mediated drug-loaded nano-microcapsules delivery system may significantly improve the efficiency of drug delivery, which may be a promising new therapeutic method.
Collapse
Affiliation(s)
- Jing Ma
- Department of Ultrasound, Shanghai First People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200080; ; Department of Cardiovascular Ultrasound, Shanghai East Hospital Affiliated to Tongji University, Shanghai 200120, P.R. China
| | - Lian Fang DU
- Department of Ultrasound, Shanghai First People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200080
| | - Ming Chen
- Department of Cardiovascular Ultrasound, Shanghai East Hospital Affiliated to Tongji University, Shanghai 200120, P.R. China
| | - Hang Hui Wang
- Department of Ultrasound, Shanghai First People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200080
| | - Ling Xi Xing
- Department of Ultrasound, Shanghai First People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200080
| | - Li Fang Jing
- Department of Ultrasound, Shanghai First People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200080
| | - Yun Hua Li
- Department of Ultrasound, Shanghai First People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200080
| |
Collapse
|
83
|
Optimization of Ultrasound-mediated Anti-angiogenic Cancer Gene Therapy. MOLECULAR THERAPY-NUCLEIC ACIDS 2013; 2:e94. [PMID: 23695537 PMCID: PMC4817934 DOI: 10.1038/mtna.2013.20] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Ultrasound-targeted microbubble destruction (UTMD) can be used to deliver silencing gene therapy to tumors. We hypothesized that UTMD would be effective in suppressing angiogenesis within tumors, and that modulation of the ultrasound pulsing intervals (PI) during UTMD would affect the magnitude of target knockdown. We performed UTMD of vascular endothelial growth factor receptor-2 (VEGFR2) short hairpin (sh)RNA plasmid in an heterotopic mammary adenocarcinoma model in rats, evaluating PIs of 2, 5, 10, and 20 seconds. We demonstrated that UTMD with a PI of 10 seconds resulted in the greatest knockdown of VEGFR2 by PCR, immunostaining, western blotting, smaller tumor volumes and perfused areas, and lower tumor microvascular blood volume (MBV) and flow by contrast-enhanced ultrasound (CEU) compared with UTMD-treated tumors at 2, 5, and 20 seconds, control tumors, tumors treated with intravenous shRNA plasmid and scrambled plasmid. CEU perfusion assessment using the therapeutic probe demonstrated that tumors were fully replenished with microbubbles within 10 seconds, but incompletely replenished at PI-2 and PI-5 seconds. In conclusion, for anti-VEGFR2 cancer gene therapy by UTMD, PI of 10 seconds results in higher target knockdown and a greater anti-angiogenic effect. Complete replenishment of tumor vasculature with silencing gene-bearing microbubbles in between destructive pulses of UTMD is required to maximize the efficacy of anti-angiogenic cancer gene therapy.Molecular Therapy - Nucleic Acids (2013) 2, e94; doi:10.1038/mtna.2013.20; published online 21 May 2013.
Collapse
|
84
|
Leong-Poi H. Contrast ultrasound and targeted microbubbles: diagnostic and therapeutic applications in progressive diabetic nephropathy. Semin Nephrol 2013; 32:494-504. [PMID: 23062991 DOI: 10.1016/j.semnephrol.2012.07.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Diabetic nephropathy remains one of the most common causes for end-stage renal disease worldwide. Although therapies aimed at optimizing glycemic control and systemic blood pressure have benefit, the reduction in progressive nephropathy remains modest at best. Thus, research continues to focus on newer therapies to address the unmet needs for additional renal protective strategies. The ability to noninvasively image the molecular and cellular processes that underlie diabetic nephropathy would be useful in risk stratifying patients with diabetes, and more importantly would aid in the evaluation of novel therapies to prevent and treat nephropathy. In addition, the development of ultrasound technologies that allow targeted gene delivery using high-power ultrasound and DNA-bearing microbubbles may have applicability for gene therapy to prevent diabetic nephropathy. This review highlights contrast-enhanced ultrasound imaging techniques for the evaluation of renal pathologies, including perfusion and molecular imaging techniques, and ultrasound-mediated gene delivery for therapeutic applications in diabetic nephropathy, that have potential for translation to clinical practice.
Collapse
Affiliation(s)
- Howard Leong-Poi
- Division of Cardiology, Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
85
|
Katz MG, Fargnoli AS, Bridges CR. Myocardial gene transfer: routes and devices for regulation of transgene expression by modulation of cellular permeability. Hum Gene Ther 2013; 24:375-92. [PMID: 23427834 DOI: 10.1089/hum.2012.241] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Heart diseases are major causes of morbidity and mortality in Western society. Gene therapy approaches are becoming promising therapeutic modalities to improve underlying molecular processes affecting failing cardiomyocytes. Numerous cardiac clinical gene therapy trials have yet to demonstrate strong positive results and advantages over current pharmacotherapy. The success of gene therapy depends largely on the creation of a reliable and efficient delivery method. The establishment of such a system is determined by its ability to overcome the existing biological barriers, including cellular uptake and intracellular trafficking as well as modulation of cellular permeability. In this article, we describe a variety of physical and mechanical methods, based on the transient disruption of the cell membrane, which are applied in nonviral gene transfer. In addition, we focus on the use of different physiological techniques and devices and pharmacological agents to enhance endothelial permeability. Development of these methods will undoubtedly help solve major problems facing gene therapy.
Collapse
Affiliation(s)
- Michael G Katz
- Thoracic and Cardiovascular Surgery, Sanger Heart & Vascular Institute, Carolinas Healthcare System, Charlotte, NC 28203, USA
| | | | | |
Collapse
|
86
|
Vlaskou D, Plank C, Mykhaylyk O. Magnetic and acoustically active microbubbles loaded with nucleic acids for gene delivery. Methods Mol Biol 2013; 948:205-41. [PMID: 23070773 DOI: 10.1007/978-1-62703-140-0_15] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Targeted gene or drug delivery aims to locally accumulate the active agent and achieve the maximum local therapeutic effect at the target-site while reducing unwanted effects at nontarget sites. A further development of the magnetic drug-targeting concept is combining it with an ultrasound-triggered delivery using magnetic microbubbles as a carrier for gene or drug delivery. For this purpose, selected magnetic nanoparticles (MNPs), phospholipids, and nucleic acid are assembled in the presence of perfluorocarbon gas into flexible formulations of magnetic lipospheres or microbubbles. This manuscript describes the protocols for preparation of magnetic lipospheres and microbubbles for nucleic acid delivery, and it also describes the procedures for labeling the components of the bubbles (lipids, MNPs, and nucleic acids) for the visualization of the vectors and their characterization, such as magnetic responsiveness and ultrasound contrast effects. Protocols are given for the transfection procedure in adherent cells, evaluation of the association of the magnetic vectors with the cells, reporter gene expression analysis, and cell viability assessment.
Collapse
Affiliation(s)
- Dialechti Vlaskou
- Institute of Experimental Oncology and Therapy Research, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.
| | | | | |
Collapse
|
87
|
Sutton JT, Haworth KJ, Pyne-Geithman G, Holland CK. Ultrasound-mediated drug delivery for cardiovascular disease. Expert Opin Drug Deliv 2013; 10:573-92. [PMID: 23448121 DOI: 10.1517/17425247.2013.772578] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
INTRODUCTION Ultrasound (US) has been developed as both a valuable diagnostic tool and a potent promoter of beneficial tissue bioeffects for the treatment of cardiovascular disease. These effects can be mediated by mechanical oscillations of circulating microbubbles, or US contrast agents, which may also encapsulate and shield a therapeutic agent in the bloodstream. Oscillating microbubbles can create stresses directly on nearby tissue or induce fluid effects that effect drug penetration into vascular tissue, lyse thrombi or direct drugs to optimal locations for delivery. AREAS COVERED The present review summarizes investigations that have provided evidence for US-mediated drug delivery as a potent method to deliver therapeutics to diseased tissue for cardiovascular treatment. In particular, the focus will be on investigations of specific aspects relating to US-mediated drug delivery, such as delivery vehicles, drug transport routes, biochemical mechanisms and molecular targeting strategies. EXPERT OPINION These investigations have spurred continued research into alternative therapeutic applications, such as bioactive gas delivery and new US technologies. Successful implementation of US-mediated drug delivery has the potential to change the way many drugs are administered systemically, resulting in more effective and economical therapeutics, and less-invasive treatments.
Collapse
Affiliation(s)
- Jonathan T Sutton
- University of Cincinnati, College of Medicine, Internal Medicine, Division of Cardiovascular Diseases, and Biomedical Engineering Program, Cincinnati, OH, USA
| | | | | | | |
Collapse
|
88
|
Vu V, Liu Y, Sen S, Xu A, Sweeney G. Delivery of adiponectin gene to skeletal muscle using ultrasound targeted microbubbles improves insulin sensitivity and whole body glucose homeostasis. Am J Physiol Endocrinol Metab 2013; 304:E168-75. [PMID: 23132298 PMCID: PMC3543570 DOI: 10.1152/ajpendo.00493.2012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Numerous studies have shown that adiponectin confers antidiabetic effects via both insulin-like and insulin-sensitizing actions. The majority of adiponectin in circulation is derived from adipocytes; however, other tissues such as skeletal muscle can produce adiponectin. This study was designed to investigate the functional significance of adiponectin produced by skeletal muscle. We encapsulated the adiponectin gene in lipid-coated microspheres filled with octafluoropropane gas that were injected into the systemic circulation and destroyed within the microvasculature of skeletal muscle using ultrasound. We first demonstrated safe and successful targeting of luciferase and green fluorescent protein reporter genes to skeletal muscle using this approach and then confirmed efficient overexpression of adiponectin mRNA and oligomeric protein forms. Glucose tolerance test indicated that overexpression of adiponectin in skeletal muscle was able to improve glucose intolerance induced by feeding mice a high-fat diet (HFD), and this correlated with improved skeletal muscle insulin signaling. We then performed hyperinsulinemic-euglycemic clamp studies and demonstrated that adiponectin overexpression attenuated the decreases in glucose infusion rate, glucose disposal, and increase in glucose appearance induced by HFD. Ultrasound-targeted microbubble destruction (UTMD) delivery of adiponectin to skeletal muscle also enhanced serum adiponectin levels and improved hepatic insulin sensitivity. In conclusion, our data show that UTMD efficiently delivers adiponectin to skeletal muscle and that this improves insulin sensitivity and glucose homeostasis.
Collapse
Affiliation(s)
- Vivian Vu
- Department of Biology, York University, Toronto, Canada
| | | | | | | | | |
Collapse
|
89
|
Sirsi SR, Borden MA. Advances in ultrasound mediated gene therapy using microbubble contrast agents. Am J Cancer Res 2012; 2:1208-22. [PMID: 23382777 PMCID: PMC3563148 DOI: 10.7150/thno.4306] [Citation(s) in RCA: 155] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Accepted: 07/01/2012] [Indexed: 12/19/2022] Open
Abstract
Microbubble ultrasound contrast agents have the potential to dramatically improve gene therapy treatments by enhancing the delivery of therapeutic DNA to malignant tissue. The physical response of microbubbles in an ultrasound field can mechanically perturb blood vessel walls and cell membranes, enhancing drug permeability into malignant tissue. In this review, we discuss literature that provided evidence of specific mechanisms that enhance in vivo gene delivery utilizing microbubble contrast agents, namely their ability to 1) improving cell membrane permeability, 2) modulate vascular permeability, and 3) enhance endocytotic uptake in cells. Additionally, we review novel microbubble vectors that are being developed in order to exploit these mechanisms and deliver higher gene payloads with greater target specificity. Finally, we discuss some future considerations that should be addressed in the development of next-generation microbubbles in order to improve in vivo microbubble gene delivery. Overall, microbubbles are rapidly gaining popularity as efficient gene carriers, and combined with their functionality as imaging contrast agents, they represent powerful theranostic tools for image guided gene therapy applications.
Collapse
|
90
|
Saliba Y, Mougenot N, Jacquet A, Atassi F, Hatem S, Farès N, Lompré AM. A new method of ultrasonic nonviral gene delivery to the adult myocardium. J Mol Cell Cardiol 2012; 53:801-8. [DOI: 10.1016/j.yjmcc.2012.07.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Revised: 06/27/2012] [Accepted: 07/26/2012] [Indexed: 10/28/2022]
|
91
|
Xie A, Belcik T, Qi Y, Morgan TK, Champaneri SA, Taylor S, Davidson BP, Zhao Y, Klibanov AL, Kuliszewski MA, Leong-Poi H, Ammi A, Lindner JR. Ultrasound-mediated vascular gene transfection by cavitation of endothelial-targeted cationic microbubbles. JACC Cardiovasc Imaging 2012; 5:1253-62. [PMID: 23236976 PMCID: PMC3682923 DOI: 10.1016/j.jcmg.2012.05.017] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Revised: 05/01/2012] [Accepted: 05/03/2012] [Indexed: 01/19/2023]
Abstract
OBJECTIVES Ultrasound-mediated gene delivery can be amplified by acoustic disruption of microbubble carriers that undergo cavitation. We hypothesized that endothelial targeting of microbubbles bearing cDNA is feasible and, through optimizing proximity to the vessel wall, increases the efficacy of gene transfection. BACKGROUND Contrast ultrasound-mediated gene delivery is a promising approach for site-specific gene therapy, although there are concerns with the reproducibility of this technique and the safety when using high-power ultrasound. METHODS Cationic lipid-shelled decafluorobutane microbubbles bearing a targeting moiety were prepared and compared with nontargeted microbubbles. Microbubble targeting efficiency to endothelial adhesion molecules (P-selectin or intercellular adhesion molecule [ICAM]-1) was tested using in vitro flow chamber studies, intravital microscopy of tumor necrosis factor-alpha (TNF-α)-stimulated murine cremaster muscle, and targeted contrast ultrasound imaging of P-selectin in a model of murine limb ischemia. Ultrasound-mediated transfection of luciferase reporter plasmid charge coupled to microbubbles in the post-ischemic hindlimb muscle was assessed by in vivo optical imaging. RESULTS Charge coupling of cDNA to the microbubble surface was not influenced by the presence of targeting ligand, and did not alter the cavitation properties of cationic microbubbles. In flow chamber studies, surface conjugation of cDNA did not affect attachment of targeted microbubbles at microvascular shear stresses (0.6 and 1.5 dyne/cm(2)). Attachment in vivo was also not affected by cDNA according to intravital microscopy observations of venular adhesion of ICAM-1-targeted microbubbles and by ultrasound molecular imaging of P-selectin-targeted microbubbles in the post-ischemic hindlimb in mice. Transfection at the site of high acoustic pressures (1.0 and 1.8 MPa) was similar for control and P-selectin-targeted microbubbles but was associated with vascular rupture and hemorrhage. At 0.6 MPa, there were no adverse bioeffects, and transfection was 5-fold greater with P-selectin-targeted microbubbles. CONCLUSIONS We conclude that ultrasound-mediated transfection at safe acoustic pressures can be markedly augmented by endothelial juxtaposition.
Collapse
Affiliation(s)
- Aris Xie
- Division of Cardiovascular Medicine, Oregon Health & Science University, Portland, Oregon
| | - Todd Belcik
- Division of Cardiovascular Medicine, Oregon Health & Science University, Portland, Oregon
| | - Yue Qi
- Division of Cardiovascular Medicine, Oregon Health & Science University, Portland, Oregon
| | - Terry K. Morgan
- Department of Pathology, Oregon Health & Science University, Portland, Oregon
| | | | - Sarah Taylor
- Cardiovascular Division, University of Virginia, Charlottesville, Virginia
| | - Brian P. Davidson
- Division of Cardiovascular Medicine, Oregon Health & Science University, Portland, Oregon
| | - Yan Zhao
- Division of Cardiovascular Medicine, Oregon Health & Science University, Portland, Oregon
| | | | | | | | - Azzdine Ammi
- Division of Cardiovascular Medicine, Oregon Health & Science University, Portland, Oregon
| | - Jonathan R. Lindner
- Division of Cardiovascular Medicine, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
92
|
Ultrasound and microbubble-assisted gene delivery: recent advances and ongoing challenges. Ther Deliv 2012; 3:1199-215. [PMID: 23116012 DOI: 10.4155/tde.12.100] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Having first been developed for ultrasound imaging, nowadays, microbubbles are proposed as tools for ultrasound-assisted gene delivery, too. Their behavior during ultrasound exposure causes transient membrane permeability of surrounding cells, facilitating targeted local delivery. The increased cell uptake of extracellular compounds by ultrasound in the presence of microbubbles is attributed to a phenomenon called sonoporation. Sonoporation has been successfully applied to deliver nucleic acids in vitro and in vivo in a variety of therapeutic applications. However, the biological and physical mechanisms of sonoporation are still not fully understood. In this review, we discuss recent data concerning microbubble--cell interactions leading to sonoporation and we report on the progress in ultrasound-assisted therapeutic gene delivery in different organs. In addition, we outline ongoing challenges of this novel delivery method for its clinical use.
Collapse
|
93
|
Chen S, Shimoda M, Chen J, Grayburn PA. Stimulation of adult resident cardiac progenitor cells by durable myocardial expression of thymosin beta 4 with ultrasound-targeted microbubble delivery. Gene Ther 2012; 20:225-33. [DOI: 10.1038/gt.2012.89] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
94
|
Tlaxca JL, Rychak JJ, Ernst PB, Konkalmatt PR, Shevchenko TI, Pizarro TT, Pizzaro TT, Rivera-Nieves J, Klibanov AL, Lawrence MB. Ultrasound-based molecular imaging and specific gene delivery to mesenteric vasculature by endothelial adhesion molecule targeted microbubbles in a mouse model of Crohn's disease. J Control Release 2012; 165:216-25. [PMID: 23142578 DOI: 10.1016/j.jconrel.2012.10.021] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Revised: 09/27/2012] [Accepted: 10/15/2012] [Indexed: 02/06/2023]
Abstract
Crohn's disease (CD) is a chronic inflammatory disorder of the gastrointestinal tract (GI) for which treatments with immunosuppressive drugs have significant side-effects. Consequently, there is a clinical need for site-specific and non-toxic delivery of therapeutic genes or drugs for CD and related disorders such as inflammatory bowel disease. The aim of this study was to validate a gene delivery platform based on ultrasound-activated lipid-shelled microbubbles (MBs) targeted to inflamed mesenteric endothelium in the CD-like TNFΔARE mouse model. MBs bearing luciferase plasmid were functionalized with antibodies to MAdCAM-1 (MB-M) or VCAM-1 (MB-V), biomarkers of gut endothelial cell inflammation and evaluated in an in vitro flow chamber assay with appropriate ligands to confirm targeting specificity. Following MB retro-orbital injection in TNFΔARE mice, the mean contrast intensity in the ileocecal region from accumulated MB-M and MB-V was 8.5-fold and 3.6-fold greater, respectively, compared to MB-C. Delivery of luciferase plasmid to the GI tract in TNFΔARE mice was achieved by insonating the endothelial cell-bound agents using a commercial sonoporator. Luciferase expression in the midgut was detected 48 h later by bioluminescence imaging and further confirmed by immunohistochemical staining. The liver, spleen, heart, and kidney had no detectable bioluminescence following insonation. Transfection of the microcirculation guided by a targeted, acoustically-activated platform such as an ultrasound contrast agent microbubble has the potential to be a minimally-invasive treatment strategy to ameliorate CD and other inflammatory conditions.
Collapse
Affiliation(s)
- José L Tlaxca
- Department of Biomedical Engineering, School of Engineering and Applied Science, University of Virginia, Charlottesville, VA, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Carson AR, McTiernan CF, Lavery L, Grata M, Leng X, Wang J, Chen X, Villanueva FS. Ultrasound-targeted microbubble destruction to deliver siRNA cancer therapy. Cancer Res 2012; 72:6191-9. [PMID: 23010078 DOI: 10.1158/0008-5472.can-11-4079] [Citation(s) in RCA: 125] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Microbubble contrast agents can specifically deliver nucleic acids to target tissues when exposed to ultrasound treatment parameters that mediate microbubble destruction. In this study, we evaluated whether microbubbles and ultrasound-targeted microbubble destruction (UTMD) could be used to enhance delivery of EGF receptor (EGFR)-directed siRNA to murine squamous cell carcinomas. Custom-designed microbubbles efficiently bound siRNA and mediated RNAse protection. UTMD-mediated delivery of microbubbles loaded with EGFR-directed siRNA to murine squamous carcinoma cells in vitro reduced EGFR expression and EGF-dependent growth, relative to delivery of control siRNA. Similarly, serial UTMD-mediated delivery of EGFR siRNA to squamous cell carcinoma in vivo decreased EGFR expression and increased tumor doubling time, relative to controls receiving EGFR siRNA-loaded microbubbles but not ultrasound or control siRNA-loaded microbubbles and UTMD. Taken together, our results offer a preclinical proof-of-concept for customized microbubbles and UTMD to deliver gene-targeted siRNA for cancer therapy.
Collapse
Affiliation(s)
- Andrew R Carson
- Center for Ultrasound Molecular Imaging and Therapeutics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | | | | | | | | | | | |
Collapse
|
96
|
Kharlamov AN, Gabinsky JL. Plasmonic photothermic and stem cell therapy of atherosclerotic plaque as a novel nanotool for angioplasty and artery remodeling. Rejuvenation Res 2012; 15:222-30. [PMID: 22533437 DOI: 10.1089/rej.2011.1305] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Some modern angioplasty techniques drastically affect the geometry of the plaque and the lumen, but have some inherent clinical and technical limitations. METHODS A total of 101 Yucatan miniature swine were allocated to the three following groups (34 pigs into 60/15- to 70/40-nm silica-gold nanoparticles (NPs), 34 swine into ferromagnetic group with iron-bearing NPs and delivery in hand of magnetic fields, and 33 in a sirolimus stenting control). Animals in the nanogroup were subdivided further into four subsets according to the delivery approach: (1) Intracoronary infused circulating stem progenitor cells (SPCs), including SP(+) (side population) cells, (2) intracoronary infused, ultrasound-mediated, albumin-coated, gas-filled microbubbles, (3) CD73(+)105(+) SPCs in the composition of a bioengineered on-artery patch (cardiac surgery), (4) CD73(+)CD105(+) SPCs engrafted by manual subadventitial injection (cardiac surgery). NPs were detonated with a microwatt near-infrared (NIR) laser (821 nm, 35-44 W/cm(2) for 7 min of exposure). RESULTS Changes of the total atheroma volume (TAV; mm(3)) immediately after the laser irradiation at month 6 in the nanoshell, ferromagnetic, and control groups were -7.54%/-22.92%, -9.7%/-16.84%, and -10.5%/-7.06% (p<0.01), respectively, and in the subsets reached -2.79%/-21.92%, -6.26%/-15.24%, -4.6%/-31.21%, -16.5%/-23.3% (p<0.05), respectively. Some cases of atherothrombosis and distal embolism (23.5%) were documented only in the microbubbles subset. The impact of the therapy on the nonorganic part of the plaque-antiinflammative and antiapoptotic effects, signs of neovascularization, and restoration of artery function-were predominant in the observed subsets with SPCs (p<0.01). CONCLUSION Nanoburning, especially in combination with stem cell technologies, is a very challenging technique for altering advanced plaque and holds the promise of revolutionizing state-of-the-art interventional cardiology, assuring destruction of plaque and functional restoration of the vessel wall. It could potentially become the current mechanical and pharmacological treatment.
Collapse
|
97
|
|
98
|
Wood SC, Antony S, Brown RP, Chen J, Gordon EA, Hitchins VM, Zhang Q, Liu Y, Maruvada S, Harris GR. Effects of ultrasound and ultrasound contrast agent on vascular tissue. Cardiovasc Ultrasound 2012; 10:29. [PMID: 22805356 PMCID: PMC3493263 DOI: 10.1186/1476-7120-10-29] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Accepted: 06/25/2012] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Ultrasound (US) imaging can be enhanced using gas-filled microbubble contrast agents. Strong echo signals are induced at the tissue-gas interface following microbubble collapse. Applications include assessment of ventricular function and virtual histology. AIM While ultrasound and US contrast agents are widely used, their impact on the physiological response of vascular tissue to vasoactive agents has not been investigated in detail. METHODS AND RESULTS In the present study, rat dorsal aortas were treated with US via a clinical imaging transducer in the presence or absence of the US contrast agent, Optison. Aortas treated with both US and Optison were unable to contract in response to phenylephrine or to relax in the presence of acetylcholine. Histology of the arteries was unremarkable. When the treated aortas were stained for endothelial markers, a distinct loss of endothelium was observed. Importantly, terminal deoxynucleotidyl transferase mediated dUTP nick-end-labeling (TUNEL) staining of treated aortas demonstrated incipient apoptosis in the endothelium. CONCLUSIONS Taken together, these ex vivo results suggest that the combination of US and Optison may alter arterial integrity and promote vascular injury; however, the in vivo interaction of Optison and ultrasound remains an open question.
Collapse
Affiliation(s)
- Steven C Wood
- Food and Drug Administration, Center for Devices and Radiological Health (CDRH), 10903 New Hampshire Avenue, Silver Spring, MD, 20993, USA
| | - Sible Antony
- Food and Drug Administration, Center for Devices and Radiological Health (CDRH), 10903 New Hampshire Avenue, Silver Spring, MD, 20993, USA
- School of Medicine and Health Sciences, The George Washington University, 2300, Eye Street, NW, Washington, DC, 20037, USA
| | - Ronald P Brown
- Food and Drug Administration, Center for Devices and Radiological Health (CDRH), 10903 New Hampshire Avenue, Silver Spring, MD, 20993, USA
| | - Jin Chen
- Food and Drug Administration, Center for Drug Evaluation and Research (CDER), 10903 New Hampshire Avenue, Silver Spring, MD, 20993, USA
| | - Edward A Gordon
- Food and Drug Administration, Center for Devices and Radiological Health (CDRH), 10903 New Hampshire Avenue, Silver Spring, MD, 20993, USA
| | - Victoria M Hitchins
- Food and Drug Administration, Center for Devices and Radiological Health (CDRH), 10903 New Hampshire Avenue, Silver Spring, MD, 20993, USA
| | - Qin Zhang
- Food and Drug Administration, Center for Devices and Radiological Health (CDRH), 10903 New Hampshire Avenue, Silver Spring, MD, 20993, USA
| | - Yunbo Liu
- Food and Drug Administration, Center for Devices and Radiological Health (CDRH), 10903 New Hampshire Avenue, Silver Spring, MD, 20993, USA
| | - Subha Maruvada
- Food and Drug Administration, Center for Devices and Radiological Health (CDRH), 10903 New Hampshire Avenue, Silver Spring, MD, 20993, USA
| | - Gerald R Harris
- Food and Drug Administration, Center for Devices and Radiological Health (CDRH), 10903 New Hampshire Avenue, Silver Spring, MD, 20993, USA
| |
Collapse
|
99
|
Huang Q, Deng J, Xie Z, Wang F, Chen S, Lei B, Liao P, Huang N, Wang Z, Wang Z, Cheng Y. Effective gene transfer into central nervous system following ultrasound-microbubbles-induced opening of the blood-brain barrier. ULTRASOUND IN MEDICINE & BIOLOGY 2012; 38:1234-1243. [PMID: 22677255 DOI: 10.1016/j.ultrasmedbio.2012.02.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Revised: 02/16/2012] [Accepted: 02/20/2012] [Indexed: 06/01/2023]
Abstract
To investigate whether ultrasound-targeted microbubble destruction (UTMD) could transfer gene into central nervous system (CNS) following blood-brain barrier disruption (BBBD), DNA-loaded microbubbles were infused into the mice intravenously following ultrasonic exposure. Opening of the BBB, changes of mRNA and expression of enhanced green fluorescent protein (EGFP), and safety evaluation were measured. By UTMD, EGFP were substantially expressed in the cytoplasm of the neurons at the sonicated area with minor erythrocytes extravasation and the mRNA and expression of EGFP were markedly enhanced by about 15-fold and 10-fold, respectively, than that with US alone (p < 0.01). No EGFP was detected in the mice treated with DNA-loaded microbubbles or plasmid alone. The gene expression reached a climax at 48 h, gradually reduced to a much lower level thereafter. These results demonstrated UTMD could effectively enhance exogenous gene delivery and expression in CNS following BBBD, and this technique may provide a new method for CNS gene therapy.
Collapse
Affiliation(s)
- Qin Huang
- Department of Neurosurgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Wang DS, Panje C, Pysz MA, Paulmurugan R, Rosenberg J, Gambhir SS, Schneider M, Willmann JK. Cationic versus neutral microbubbles for ultrasound-mediated gene delivery in cancer. Radiology 2012; 264:721-32. [PMID: 22723497 DOI: 10.1148/radiol.12112368] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
PURPOSE To test whether plasmid-binding cationic microbubbles (MBs) enhance ultrasound-mediated gene delivery efficiency relative to control neutral MBs in cell culture and in vivo tumors in mice. MATERIALS AND METHODS Animal studies were approved by the institutional animal care committee. Cationic and neutral MBs were characterized in terms of size, charge, circulation time, and DNA binding. Click beetle luciferase (CBLuc) reporter plasmids were mixed with cationic or neutral MBs. The ability of cationic MBs to protect bound plasmids from nuclease degradation was tested by means of a deoxyribonuclease (DNase) protection assay. Relative efficiencies of ultrasound-mediated transfection (ultrasound parameters: 1 MHz, 1 W/cm(2), 20% duty cycle, 1 minute) of CBLuc to endothelial cells by using cationic, neutral, or no MBs were compared in cell culture. Ultrasound-mediated gene delivery to mouse hind limb tumors was performed in vivo (n = 24) with insonation (1 MHz, 2 W/cm(2), 50% duty cycle, 5 minutes) after intravenous administration of CBLuc with cationic, neutral, or no MBs. Tumor luciferase activity was assessed by means of serial in vivo bioluminescence imaging and ex vivo analysis. Results were compared by using analysis of variance. RESULTS Cationic MBs (+15.8 mV; DNA binding capacity, 0.03 pg per MB) partially protected bound DNA from DNase degradation. Mean CBLuc expression of treated endothelial cells in culture was 20-fold higher with cationic than with neutral MBs (219.0 relative light units [RLUs]/µg protein ± 92.5 [standard deviation] vs 10.9 RLUs/µg protein ± 2.7, P = .001) and was significantly higher (P < .001) than that in the no MB and no ultrasound control groups. Serial in vivo bioluminescence of mouse tumors was significantly higher with cationic than with neutral MBs ([5.9 ± 2.2] to [9.3 ± 5.2] vs [2.4 ± 0.8] to [2.9 ± 1.1] × 10(4) photons/sec/cm(2)/steradian, P < .0001) and versus no MB and no ultrasound controls (P < .0001). Results of ex vivo analysis confirmed these results (ρ = 0.88, P < .0001). CONCLUSION Plasmid-binding cationic MBs enhance ultrasound-mediated gene delivery efficiency relative to neutral MBs in both cell culture and mouse hind limb tumors.
Collapse
Affiliation(s)
- David S Wang
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, 300 Pasteur Dr, Room H1307, Stanford, CA 94305, USA
| | | | | | | | | | | | | | | |
Collapse
|